AU2021270095A1 - 1, 4, 5, 6-tetrahydropyrimidine-2-amine derivative - Google Patents

1, 4, 5, 6-tetrahydropyrimidine-2-amine derivative Download PDF

Info

Publication number
AU2021270095A1
AU2021270095A1 AU2021270095A AU2021270095A AU2021270095A1 AU 2021270095 A1 AU2021270095 A1 AU 2021270095A1 AU 2021270095 A AU2021270095 A AU 2021270095A AU 2021270095 A AU2021270095 A AU 2021270095A AU 2021270095 A1 AU2021270095 A1 AU 2021270095A1
Authority
AU
Australia
Prior art keywords
group
fluoro
indazole
amino
tetrahydropyrimidin
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Pending
Application number
AU2021270095A
Inventor
Yasuhiro Aga
Toru Hasegawa
Ken-Ichi Komori
Takashi Matsushita
Ayumi Ogawa
Hayato Shimizu
Yasunori Tokunaga
Haruka YAMADA
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Ube Corp
Original Assignee
Ube Corp
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Ube Corp filed Critical Ube Corp
Publication of AU2021270095A1 publication Critical patent/AU2021270095A1/en
Pending legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/535Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with at least one nitrogen and one oxygen as the ring hetero atoms, e.g. 1,2-oxazines
    • A61K31/53751,4-Oxazines, e.g. morpholine
    • A61K31/53771,4-Oxazines, e.g. morpholine not condensed and containing further heterocyclic rings, e.g. timolol
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/04Drugs for skeletal disorders for non-specific disorders of the connective tissue
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • A61P3/08Drugs for disorders of the metabolism for glucose homeostasis
    • A61P3/10Drugs for disorders of the metabolism for glucose homeostasis for hyperglycaemia, e.g. antidiabetics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • A61P35/04Antineoplastic agents specific for metastasis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/02Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings
    • C07D403/12Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing two hetero rings linked by a chain containing hetero atoms as chain links
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D403/00Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00
    • C07D403/14Heterocyclic compounds containing two or more hetero rings, having nitrogen atoms as the only ring hetero atoms, not provided for by group C07D401/00 containing three or more hetero rings

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • General Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Diabetes (AREA)
  • Epidemiology (AREA)
  • Biomedical Technology (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Immunology (AREA)
  • Oncology (AREA)
  • Endocrinology (AREA)
  • Hematology (AREA)
  • Obesity (AREA)
  • Cardiology (AREA)
  • Heart & Thoracic Surgery (AREA)
  • Emergency Medicine (AREA)
  • Neurology (AREA)
  • Neurosurgery (AREA)
  • Ophthalmology & Optometry (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)

Abstract

The present invention provides a compound, or a pharmaceutically-acceptable salt thereof, represented by general formula (I) or (II) below [wherein A is a C

Description

221102_G2572WO English Translation (Final) clean copy
Description
Title of Invention:
1,4,5,6-TETRAHYDROPYRIMIDIN-2-AMINE DERIVATIVE
Technical Field
[0001] The present invention relates to a 1,4,5,6
tetrahydropyrimidin-2-amine derivative and a
pharmaceutical composition containing the same and
particularly relates to a 1,4,5,6-tetrahydropyrimidin-2
amine derivative for the prevention, alleviation and/or
remedy of a disease related to integrin av and a
pharmaceutical composition containing the same.
Background Art
[0002] In recent years, integrins have received attention
in terms of possessing cell adhesion or intracellular
signal transduction functions in response to
extracellular matrix information. The integrins are
proteins that are expressed on cell surface, and are
composed of a heterodimer of two subunits u and $ chains.
24 integrins form some subfamilies by combinations of 18
u chains and 8 $ chains. RGD integrin, one of the major
subfamilies, recognizes a ligand containing a RGD
(arginine-glycine-aspartic acid) motif in a protein
sequence. The RGD integrin includes 8 subtypes axvf1,
221102_G2572WO English Translation (Final) clean copy
avP3, avP5, avf6, avf8, axIIb3, a5f1, and u81 (Non
Patent Literature 1).
[0003] Integrin av is known to participate in various
diseases, and its inhibition is expected to have a
therapeutic effect on, for example, cardiovascular
disease, diabetic disease, neurodegenerative disease,
cancer disease, tumor metastasis, eye disease, autoimmune
disease, bone disease, and various fibroses (Non Patent
Literatures 2 to 6).
[0004] Transforming growth factor $ (TGF-) is known to
control cell proliferation, differentiation, or the like
and to participate in diseases such as fibrosis,
autoimmune disease, and cancer, and is secreted as an
inactive complex with latency-associated protein (LAP)
(Non Patent Literature 7). In research using cell, it
has been reported that RGD integrin such as integrin
axvpl, avP3, avP5, axvf6 and av8 activates latent TGF
(Non Patent Literatures 8 and 9).
[0005] Fibrosis, a pathological feature of many diseases,
is caused by the dysfunction of body's original ability
to repair injured tissues. Fibrosis brings about
excessive scarring that surpasses the wound healing
response of an important organ, and might cause
unrecoverable injury and final organ failure (Non Patent
Literature 10). In fibrosis, extracellular matrix is
mainly produced by activated fibroblasts
(myofibroblasts), and TGF- is known to participate in
221102_G2572WO English Translation (Final) clean copy
the activation of the fibroblasts (Non Patent Literature
11).
[0006] The expression of integrins also participates in
fibrosis. For example, the expression level of integrin
$5 or P6 markedly increases after tissue injury and plays
an important in vivo role in tissue fibrosis (Non Patent
Literatures 12 and 13). The increased expression level
of integrin $6 is also related to increase in the death
rate of fibrosis patients (Non Patent Literature 14).
[0007] The inhibition of integrins is known to exhibit
resistance to various fibroses. For example, integrin
$6-knockout mice inhibit the activation of TGF- and
exhibit resistance to various fibroses (Non Patent
Literatures 15 and 16). It has also been reported that
fibroblast-specific deficiency in integrin av similarly
exhibits resistance to various fibroses (Non Patent
Literatures 17 and 18).
[0008] For example, pirfenidone and nintedanib for
idiopathic pulmonary fibrosis (IPF) are used as
therapeutic drugs for fibrosis. However, any safe and
established therapeutic drug for various organ fibroses
has not yet been found. Thus, there is a demand for a
therapeutic drug for fibrosis that is effective for
various organs with reduced adverse reactions.
[0009] Research has been made so far on integrins
involved in the functional control of various cells or
221102_G2572WO English Translation (Final) clean copy
their inhibitors (Patent Literatures 1 to 8 and Non
Patent Literature 19).
Citation List
Patent Literature
[0010]
Patent Literature 1: WO 1997/008145
Patent Literature 2: WO 1999/044994
Patent Literature 3: WO 1999/052896
Patent Literature 4: WO 2000/051686
Patent Literature 5: WO 2004/060376
Patent Literature 6: WO 2014/015054
Patent Literature 7: US 2014/0051715
Patent Literature 8: WO 2017/117538
Non Patent Literature
[0011]
Non Patent Literature 1: Cell, 2002, 110 (6), 673-687
Non Patent Literature 2: Cellular and Molecular Life
Sciences, 2017, 74 (12), 2263-2282
Non Patent Literature 3: Pharmacology Research and
Perspectives, 2017, 5 (5), e00354
Non Patent Literature 4: Neurobiology of Aging, 2008, 29
(10), 1485-1493
Non Patent Literature 5: Angewandte Chemie International
Edition in English, 2018, 57 (13), 3298-3321
Non Patent Literature 6: Clinical & Experimental
Metastasis, 2003, 20, 203-213
221102_G2572WO English Translation (Final) clean copy
Non Patent Literature 7: Cold Spring Harbor Perspectives
in Biology, 2016, 8 (5), a021873
Non Patent Literature 8: Cold Spring Harbor Perspectives
in Biology, 2011, 3 (11), a005017
Non Patent Literature 9: Science Translational Medicine,
2015, 7 (288), 288ra79
Non Patent Literature 10: Molecular Aspects of Medicine,
2019, 65, 2-15
Non Patent Literature 11: Nature Reviews Nephrology,
2016, 12 (6), 325-338
Non Patent Literature 12: Journal of Hepatology, 2008, 48
(3), 453-464
Non Patent Literature 13: The American Journal of
Pathology, 2004, 164 (4), 1275-1292
Non Patent Literature 14: European Respiratory Journal,
2015, 46 (2), 486-494
Non Patent Literature 15: The American Journal of
Pathology, 2003, 163 (4), 1261-1273
Non Patent Literature 16: Cell, 1999, 96 (3), 319-328
Non Patent Literature 17: Nature Medicine, 2013, 19 (12),
1617-1624
Non Patent Literature 18: Nature Communications, 2017, 8
(1), 1118
Non Patent Literature 19: Biochemical Pharmacology, 2016,
117, 88-96
Summary of Invention
221102_G2572WO English Translation (Final) clean copy
Technical Problem
[0012] The present invention relates to the provision of
a novel compound useful in the prevention, alleviation
and/or remedy of a disease related to integrin av and a
pharmaceutical composition containing the same.
Solution to Problem
[0013] The present inventors have conducted diligent
studies on compounds having integrin av inhibitory
activity and consequently found that a series of 1,4,5,6
tetrahydropyrimidin-2-amine derivatives having an
intramolecular indazole structure, or pharmaceutically
acceptable salts thereof have excellent integrin av
(av$l, av$6, etc.) inhibitory activity, excellent
pharmacokinetic characteristics or excellent safety. The
1,4,5,6-tetrahydropyrimidin-2-amine derivative according
to the present invention or a pharmaceutically acceptable
salt thereof can be useful in the prevention, alleviation
and/or remedy of a disease related to integrin av,
preferably the prevention, alleviation and/or remedy of
fibrosis.
[0014] Specifically, the present invention is as follows.
[1] A compound represented by the following general
formula (I) or (II) or a pharmaceutically acceptable salt
thereof:
221102_G2572WO English Translation (Final) clean copy
Y Y
HN N O HN N O HN N N C2R HN N CO 2 R 1 0 A 0 O A R 1x/ R N-NH HN-N
(I) (II) wherein
A is a C6-C10 aryl group or a heteroaryl group,
wherein at least one hydrogen atom of the aryl group or
the heteroaryl group is optionally replaced with a
substituent selected from the group consisting of a
halogen atom, a hydroxy group, a C1-C6 alkyl group, a C1
C6 haloalkyl group, a C2-C6 alkenyl group, a C2-C6 alkynyl
group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a
C3-C6 cycloalkyl group, a C3-C6 cycloalkenyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, a heteroaryl
group optionally substituted by a C1-C6 alkyl group, a
cyano group, a carboxyl group, a carbamoyl group, a C1-C6
alkoxycarbonyl group, a C1-C6 alkylsulfanyl group, and a
C1-C6 alkylsulfonyl group,
R is a hydrogen atom or a C1-C6 alkyl group,
R' is a hydrogen atom or a halogen atom, and
Y is a hydrogen atom, a fluorine atom or a hydroxy
group.
[2] A compound represented by the following general
formula (I) or a pharmaceutically acceptable salt
thereof:
221102_G2572WO English Translation (Final) clean copy
Y
HN N HN N CO 2 R
R 1 H0 A N-NH
(I) wherein
A is a C6-C10 aryl group or a heteroaryl group,
wherein at least one hydrogen atom of the aryl group or
the heteroaryl group is optionally replaced with a
substituent selected from the group consisting of a
halogen atom, a hydroxy group, a C1-C6 alkyl group, a C1
C6 haloalkyl group, a C2-C6 alkenyl group, a C2-C6 alkynyl
group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a
C3-C6 cycloalkyl group, a C3-C6 cycloalkenyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, a heteroaryl
group optionally substituted by a C1-C6 alkyl group, a
cyano group, a carboxyl group, a carbamoyl group, a C1-C6
alkoxycarbonyl group, a C1-C6 alkylsulfanyl group, and a
C1-C6 alkylsulfonyl group,
R is a hydrogen atom or a C1-C6 alkyl group,
R' is a hydrogen atom or a halogen atom, and
Y is a hydrogen atom, a fluorine atom or a hydroxy
group.
[3] A compound represented by the following general
formula (II) or a pharmaceutically acceptable salt
thereof:
221102_G2572WO English Translation (Final) clean copy
Y
HN N HN O' N N CO2 R 0 A
HN-N
wherein
A is a C6-C10 aryl group or a heteroaryl group,
wherein at least one hydrogen atom of the aryl group or
the heteroaryl group is optionally replaced with a
substituent selected from the group consisting of a
halogen atom, a hydroxy group, a C1-C6 alkyl group, a C1
C6 haloalkyl group, a C2-C6 alkenyl group, a C2-C6 alkynyl
group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a
C3-C6 cycloalkyl group, a C3-C6 cycloalkenyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, a heteroaryl
group optionally substituted by a C1-C6 alkyl group, a
cyano group, a carboxyl group, a carbamoyl group, a C1-C6
alkoxycarbonyl group, a C1-C6 alkylsulfanyl group, and a
C1-C6 alkylsulfonyl group,
R is a hydrogen atom or a C1-C6 alkyl group,
R' is a hydrogen atom or a halogen atom, and
Y is a hydrogen atom, a fluorine atom or a hydroxy
group.
[4] The compound according to any one of [1] to [3] or a
pharmaceutically acceptable salt thereof, wherein A is a
phenyl group, wherein
221102_G2572WO English Translation (Final) clean copy
at least one hydrogen atom of the phenyl group is
optionally replaced with a substituent selected from the
group consisting of a halogen atom, a hydroxy group, a
C1-C6 alkyl group, a C1-C6 haloalkyl group, a C2-C6 alkenyl
group, a C2-C6 alkynyl group, a C1-C alkoxy group, a C1-C6
haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkenyl group, a C3-C6 cycloalkoxy group, a
heterocyclyl group, a heteroaryl group optionally
substituted by a C1-C alkyl group, a cyano group, a
carboxyl group, a carbamoyl group, a C1-C6 alkoxycarbonyl
group, a C1-C6 alkylsulfanyl group, and a C1-C6
alkylsulfonyl group.
[5] The compound according to any one of [1] to [4] or a
pharmaceutically acceptable salt thereof, wherein A is a
phenyl group, wherein
at least one hydrogen atom of the phenyl group is
optionally replaced with a substituent selected from the
group consisting of a halogen atom, a hydroxy group, a
C1-C6 alkyl group, a C1-C6 haloalkyl group, a C2-C6 alkenyl
group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a
C3-C6 cycloalkyl group, a C3-C6 cycloalkenyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, a heteroaryl
group optionally substituted by a C1-C6 alkyl group, a
cyano group, a carboxyl group, a carbamoyl group, a C1-C6
alkoxycarbonyl group, a C1-C6 alkylsulfanyl group, and a
C1-C6 alkylsulfonyl group.
221102_G2572WO English Translation (Final) clean copy
[6] The compound according to any one of [1] to [5] or a
pharmaceutically acceptable salt thereof, wherein A is a
phenyl group, wherein
at least one hydrogen atom of the phenyl group is
optionally replaced with a substituent selected from the
group consisting of a halogen atom, a C1-C6 alkyl group,
a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a C1-C6
haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, and a heteroaryl
group optionally substituted by a C1-C6 alkyl group.
[7] The compound according to any one of [1] to [6] or a
pharmaceutically acceptable salt thereof, wherein Y is a
fluorine atom.
[8] The compound according to any one of [1] to [7] or a
pharmaceutically acceptable salt thereof, wherein
A is a group represented by the following formula
(i): *
R5 4R
0) wherein
R 2 is a hydrogen atom or a halogen atom,
R 3 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
C1-C6 haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
221102_G2572WO English Translation (Final) clean copy
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group optionally substituted by a C1-C6 alkyl group,
R4 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, or a
C1-C6 haloalkoxy group,
R5 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
C1-C6 haloalkoxy group, or a heteroaryl group optionally
substituted by a C1-C6 alkyl group, and
R 6 is a hydrogen atom or a halogen atom.
[9] The compound according to [8] or a pharmaceutically
acceptable salt thereof, wherein
R 2 is a hydrogen atom,
R3 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
C1-C6 haloalkoxy group, or a heteroaryl group optionally
substituted by a C1-C6 alkyl group,
R 4 is a hydrogen atom or a halogen atom,
R5 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, or a
C1-C6 haloalkoxy group, and
R 6 is a hydrogen atom or a halogen atom.
[10] The compound according to [1] or a pharmaceutically
acceptable salt thereof, wherein the compound is selected
from the group consisting of
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-(trifluoromethyl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
iodo-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
methyl-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
methoxy-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
fluoro-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(3,5-bis(trifluoromethyl)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(1H-pyrazol-1-yl)-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(2
fluoro-3-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-chloro-3-(trifluoromethyl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(2
fluoro-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
fluoro-3-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
methyl-3-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(3-chloro-4-fluoro-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-4-methoxyphenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3,5-dichlorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-chlorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3,5-dibromophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-iodophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-chloro-5-iodophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(5-chloro-2-fluorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(5-bromo-2-fluorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(difluoromethoxy)-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3-chloro-5-(difluoromethoxy)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-(difluoromethoxy)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(difluoromethoxy)-4-fluorophenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(5-(difluoromethoxy)-2-fluorophenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(difluoromethoxy)-5-(1H-pyrazol-1
yl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3,5-bis(difluoromethoxy)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-chloro-5-(trifluoromethoxy)phenyl)-3-(2
(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-(trifluoromethoxy)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(2
fluoro-5-(trifluoromethoxy)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
fluoro-3-(trifluoromethoxy)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
(trifluoromethoxy)phenyl)propanoic acid,
(3S)-3-(3-(difluoromethoxy)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(difluoromethyl)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-cyclopropylphenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-cyclopropoxyphenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-chlorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3-bromophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(3,5-dimethyl-1H-pyrazol-1-yl)phenyl)-3
(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)
1H-indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
morpholinophenyl)propanoic acid,
(3S)-3-(4-(difluoromethoxy)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(3
fluoro-4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2
yl)amino)-1H-indazole-6-carboxamido)acetamido)propanoic
acid,
(3S)-3-(2-(3-chloro-4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(3-chloro-5
(trifluoromethyl)phenyl)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
((5-hydroxy-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
((1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(6
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-4-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-(trifluoromethyl)phenyl)-3-(2-(6
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-4-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-4-carboxamido)acetamido)-3-(3
iodo-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-4-carboxamido)acetamido)-3-(4
fluoro-3-(trifluoromethyl)phenyl)propanoic acid, and
(3S)-3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-4-carboxamido)acetamido)-3-(3
(trifluoromethyl)phenyl)propanoic acid.
[0015] In another embodiment, the present invention
relates to a pharmaceutical composition comprising a
compound according to any one of [1] to [10] or a
pharmaceutically acceptable salt thereof, and
in an embodiment of the pharmaceutical composition,
221102_G2572WO English Translation (Final) clean copy
relates to a pharmaceutical composition for
preventing, alleviating and/or treating a disease related
to integrin av or a disease that is improved by
inhibiting integrin av, or
a pharmaceutical composition for treating fibrosis.
[0016] In an alternative embodiment, the present
invention relates to a pharmaceutical composition, more
preferably a pharmaceutical composition for the treatment
of fibrosis, i.e., the prevention, alleviation and/or
remedy of fibrosis, comprising a compound according to
any one of [1] to [10] or a pharmaceutically acceptable
salt thereof.
[0017] In an alternative embodiment, the present
invention relates to a method for the treatment of a
disease related to integrin av or a disease that is
improved by inhibiting integrin av, for example,
fibrosis, comprising administering a compound according
to any one of [1] to [10] or a pharmaceutically
acceptable salt thereof to a subject.
[0018] In an alternative embodiment, the present
invention relates to use of a compound according to any
one of [1] to [10] or a pharmaceutically acceptable salt
thereof in the treatment of a disease related to integrin
axv or a disease that is improved by inhibiting integrin
axv, for example, fibrosis.
[0019] In an alternative embodiment, the present
invention relates to the compound according to any one of
221102_G2572WO English Translation (Final) clean copy
[1] to [10] or a pharmaceutically acceptable salt thereof
for use in the treatment of a disease related to integrin
axv or a disease that is improved by inhibiting integrin
axv, for example, fibrosis.
[0020] In an alternative embodiment, the present
invention relates to use of a compound according to any
one of [1] to [10] or a pharmaceutically acceptable salt
thereof in the production of a medicament for the
treatment of a disease related to integrin av or a
disease that is improved by inhibiting integrin av, for
example, fibrosis.
[0021] In an alternative embodiment, the present
invention relates to an intermediate of a compound
according to any one of [1] to [10] or a pharmaceutically
acceptable salt thereof.
[0022] In an alternative embodiment, the present
invention relates to a method for producing a compound
according to any one of [1] to [10] or a pharmaceutically
acceptable salt thereof.
Advantageous Effects of Invention
[0023] The present invention provides a novel compound
and a method for synthesizing the compound. Another
aspect of the present invention provides a pharmaceutical
composition for the prevention, alleviation and/or remedy
of a disease related to at least any integrin av (e.g.,
fibrosis), comprising the compound, the compound for
221102_G2572WO English Translation (Final) clean copy
preventing, alleviating and/or treating the disease, or a
method for preventing, alleviating and/or treating the
disease with the compound. An alternative aspect of the
present invention provides a novel intermediate for
synthesizing the compound.
Description of Embodiments
[0024] <Definition>
In the present specification, various substituents
are as follows.
The "C6-C10 aryl group" means a monocyclic or
dicyclic monovalent aromatic hydrocarbon group having 6
to 10 ring-forming carbon atoms. Examples of the aryl
group include phenyl and naphthyl.
[0025] The "heteroaryl group" means a monovalent aromatic
heterocyclic group containing at least one heteroatom
selected from a nitrogen atom, an oxygen atom and a
sulfur atom and is preferably a monovalent aromatic
heterocyclic group having 5 to 10 ring-forming atoms.
Examples of the heteroaryl group include: monocyclic
heteroaryl groups such as pyrrolyl, furyl, thienyl,
pyrazolyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl,
isothiazolyl, triazolyl, oxadiazolyl, thiadiazolyl,
tetrazolyl, pyranyl, pyridyl, pyridazinyl, pyrimidinyl,
and pyrazinyl; and dicyclic heteroaryl groups such as
indolyl, indazolyl, benzotriazolyl, benzofuranyl,
221102_G2572WO English Translation (Final) clean copy
benzimidazolyl, benzothiophenyl, benzisoxazolyl,
benzopyranyl, benzothienyl, quinolyl, and isoquinolyl.
[0026] The "halogen" or the "halo" means a halogen atom.
Examples thereof include a fluorine atom (fluoro), a
chlorine atom (chloro), a bromine atom (bromo) and an
iodine atom (iodo).
[0027] The "C1-C6 alkyl group" means an alkyl group having
1 to 6 carbon atoms, and the alkyl group may be a linear
alkyl group or may be a branched alkyl group. Examples
of the alkyl group include methyl, ethyl, n-propyl,
isopropyl, n-butyl, isobutyl, sec-butyl, tert-butyl, n
pentyl, isopentyl, neopentyl, tert-pentyl, n-hexyl,
isohexyl, neohexyl, and tert-hexyl. The alkyl group may
be a C1-C4 alkyl group.
[0028] The "C1-C6 haloalkyl group" means a group in which
at least one hydrogen of the "C1-C6 alkyl group" is
replaced with a halogen atom. The haloalkyl group may be
a linear haloalkyl group or may be a branched haloalkyl
group. Examples of the haloalkyl group include
fluoromethyl, chloromethyl, bromomethyl, iodomethyl,
difluoromethyl, dichloromethyl, dibromomethyl,
diiodomethyl, trifluoromethyl, trichloromethyl,
tribromomethyl, triiodomethyl, 2-fluoroethyl, 2
chloroethyl, 2-bromoethyl, 2-iodoethyl, 2,2
difluoroethyl, 2,2-dichloroethyl, 2,2-dibromoethyl, 2,2
diiodoethyl, 2,2,2-trifluoroethyl, 2,2,2-trichloroethyl,
2,2,2-tribromoethyl, 2,2,2-triiodoethyl, 3-fluoropropyl,
221102_G2572WO English Translation (Final) clean copy
3-chloropropyl, 3-bromopropyl, 3-iodopropyl, 4
fluorobutyl, 4-chlorobutyl, 4-bromobutyl, 4-iodobutyl, 5
fluoropentyl, 5-chloropentyl, 5-bromopentyl, 5
iodopentyl, 6-fluorohexyl, 6-chlorohexyl, 6-bromohexyl,
and 6-iodohexyl. The haloalkyl group may be a C1-C4
haloalkyl group.
[0029] The "C2-C6 alkenyl group" means an alkenyl group
having 2 to 6 carbon atoms, and the alkenyl group may be
a linear alkenyl group or may be a branched alkenyl
group. Examples of the alkenyl group include vinyl,
allyl, 1-propenyl, 2-propenyl, 1-butenyl, 2-butenyl, 3
butenyl, 1-methyl-1-propenyl, 1-methyl-2-propenyl, 2
methyl-1-propenyl, 2-methyl-2-propenyl, 1-pentenyl, 2
pentenyl, 3-pentenyl, 4-pentenyl, 1-methyl-2-butenyl, 3
methyl-1-butenyl, 1-hexenyl, 2-hexenyl, 3-hexenyl, 4
hexenyl, 5-hexenyl, 1-methyl-2-pentenyl, and 3-methyl-1
pentenyl. The alkenyl group may be a C2-C4 alkenyl group.
[0030] The "C2-C6 alkynyl group" means an alkynyl group
having 2 to 6 carbon atoms, and the alkynyl group may be
a linear alkynyl group or may be a branched alkynyl
group. Examples of the alkynyl group include ethynyl, 1
propynyl, 2-propynyl, 1-butynyl, 2-butynyl, 3-butynyl, 1
methyl-2-propynyl, 1,1-dimethyl-2-propynyl, 1-pentynyl,
2-pentynyl, 3-pentynyl, 4-pentynyl, 1-methyl-2-butynyl,
3-methyl-1-butynyl, 1-hexynyl, 2-hexynyl, 3-hexynyl, 4
hexynyl, 5-hexynyl, 1-methyl-2-pentynyl, and 3-methyl-1
pentynyl. The alkynyl group may be a C2-C4 alkynyl group.
221102_G2572WO English Translation (Final) clean copy
[0031] The "C1-C6 alkoxy group" means a group in which the
"C1-C6 alkyl group" is bonded via an oxygen atom. The
alkoxy group may be a linear alkoxy group or may be a
branched alkoxy group. Examples of the alkoxy group
include methoxy, ethoxy, n-propoxy, isopropoxy, n-butoxy,
isobutoxy, sec-butoxy, tert-butoxy, n-pentoxy,
isopentoxy, neopentoxy, tert-pentoxy, n-hexoxy,
isohexoxy, neohexoxy, and tert-hexoxy. The alkoxy group
may be a C1-C4 alkoxy group.
[0032] The "C1-C6 haloalkoxy group" means a group in which
at least one hydrogen of the "C1-C6 alkoxy group" is
replaced with a halogen atom. The haloalkoxy group may
be a linear haloalkoxy group or may be a branched
haloalkoxy group. Examples of the haloalkoxy group
include fluoromethoxy, chloromethoxy, bromomethoxy,
iodomethoxy, difluoromethoxy, dichloromethoxy,
dibromomethoxy, diiodomethoxy, trifluoromethoxy,
trichloromethoxy, tribromomethoxy, triiodomethoxy, 2
fluoroethoxy, 2-chloroethoxy, 2-bromoethoxy, 2
iodoethoxy, 2,2-difluoroethoxy, 2,2-dichloroethoxy, 2,2
dibromoethoxy, 2,2-diiodoethoxy, 2,2,2-trifluoroethoxy,
2,2,2-trichloroethoxy, 2,2,2-tribromoethoxy, 2,2,2
triiodoethoxy, 3-fluoropropoxy, 3-chloropropoxy, 3
bromopropoxy, 3-iodopropoxy, 4-fluorobutoxy, 4
chlorobutoxy, 4-bromobutoxy, 4-iodobutoxy, 5
fluoropentoxy, 5-chloropentoxy, 5-bromopentoxy, 5
iodopentoxy, 6-fluorohexoxy, 6-chlorohexoxy, 6
221102_G2572WO English Translation (Final) clean copy
bromohexoxy, and 6-iodohexoxy. The haloalkoxy group may
be a C1-C4 haloalkoxy group.
[0033] The "C3-C6 cycloalkyl group" means an alicyclic
saturated hydrocarbon group having 3 to 6 ring-forming
carbon atoms. Examples of the cycloalkyl group include
cyclopropyl, cyclobutyl, cyclopentyl, and cyclohexyl.
[0034] The "C3-C6 cycloalkenyl group" means an alicyclic
unsaturated hydrocarbon group having 3 to 6 ring-forming
carbon atoms and having at least one carbon-carbon double
bond in the ring. Examples of the cycloalkenyl group
include cyclopropenyl, cyclobutenyl, cyclopentenyl, and
cyclohexenyl (2-cyclohexenyl and 3-cyclohexenyl).
[0035] The "C3-C6 cycloalkoxy group" means a group in
which the "C3-C6 cycloalkyl group" is bonded via an
oxygen atom. Examples of the cycloalkoxy group include
cyclopropoxy, cyclobutoxy, cyclopentoxy, and cyclohexoxy.
[0036] The "heterocyclyl group" means a saturated
monovalent heterocyclic group or a nonaromatic
unsaturated monovalent heterocyclic group containing at
least one heteroatom selected from a nitrogen atom, an
oxygen atom and a sulfur atom, and is preferably a
saturated monovalent heterocyclic group having 5 to 10
ring-forming atoms. Examples of the heterocyclyl group
include azetidinyl, oxetanyl, thietanyl, pyrrolidinyl,
piperidinyl, tetrahydrofuryl, tetrahydropyranyl,
tetrahydrothienyl (i.e., thiolanyl), piperazinyl, and
morpholinyl.
221102_G2572WO English Translation (Final) clean copy
[0037] The "C1-C6 alkoxycarbonyl group" means a group in
which the "C1-C6 alkoxy group" is bonded via a carbonyl
group. The alkoxycarbonyl group may be a linear
alkoxycarbonyl group or may be a branched alkoxycarbonyl
group. Examples of the alkoxycarbonyl group include
methoxycarbonyl, ethoxycarbonyl, n-propoxycarbonyl,
isopropoxycarbonyl, n-butoxycarbonyl, isobutoxycarbonyl,
sec-butoxycarbonyl, tert-butoxycarbonyl, n
pentoxycarbonyl, isopentoxycarbonyl, neopentoxycarbonyl,
tert-pentoxycarbonyl, n-hexoxycarbonyl,
isohexoxycarbonyl, neohexoxycarbonyl, and tert
hexoxycarbonyl. The alkoxycarbonyl group may be a C1-C4
alkoxycarbonyl group.
[0038] The "C1-C6 alkylsulfanyl group" means a group in
which the "C1-C6 alkyl group" is bonded via a sulfur
atom. The alkylsulfanyl group may be a linear
alkylsulfanyl group or may be a branched alkylsulfanyl
group. Examples of the alkylsulfanyl group include
methylsulfanyl, ethylsulfanyl, n-propylsulfanyl,
isopropylsulfanyl, n-butylsulfanyl, isobutylsulfanyl,
sec-butylsulfanyl, tert-butylsulfanyl, n-pentylsulfanyl,
isopentylsulfanyl, neopentylsulfanyl, tert
pentylsulfanyl, n-hexylsulfanyl, isohexylsulfanyl,
neohexylsulfanyl, and tert-hexylsulfanyl. The
alkylsulfanyl group may be a C1-C4 alkylsulfanyl group.
[0039] The "C1-C6 alkylsulfonyl group" means a group in
which the "C1-C6 alkyl group" is bonded via a sulfonyl
221102_G2572WO English Translation (Final) clean copy
group. The alkylsulfonyl group may be a linear
alkylsulfonyl group or may be a branched alkylsulfonyl
group. Examples of the alkylsulfonyl group include
methylsulfonyl, ethylsulfonyl, n-propylsulfonyl,
isopropylsulfonyl, n-butylsulfonyl, isobutylsulfonyl,
sec-butylsulfonyl, tert-butylsulfonyl, n-pentylsulfonyl,
isopentylsulfonyl, neopentylsulfonyl, tert
pentylsulfonyl, n-hexylsulfonyl, isohexylsulfonyl,
neohexylsulfonyl, and tert-hexylsulfonyl. The
alkylsulfonyl group may be a C1-C4 alkylsulfonyl group.
[0040] In the present specification, the
"pharmaceutically acceptable salt" includes a
pharmaceutically acceptable acid-addition salt and a salt
with a pharmaceutically acceptable acid (hereinafter,
these are also collectively referred to as an "acid
addition salt, etc."), and a pharmaceutically acceptable
base-addition salt and a salt with a pharmaceutically
acceptable base (hereinafter, these are also collectively
referred to as a "base-addition salt, etc.").
[0041] In the present specification, examples of the
"acid-addition salt" or the "salt with an acid" (acid
addition salt, etc.) include hydrochloride, hydrobromide,
sulfate, nitrate, phosphate, acetate, oxalate, malonate,
fumarate, maleate, phthalate, trifluoroacetate,
methanesulfonate, benzenesulfonate, p-toluenesulfonate,
2,4-dimethylbenzenesulfonate, 2,4,6
221102_G2572WO English Translation (Final) clean copy
trimethylbenzenesulfonate, 4-ethylbenzenesulfonate, and
naphthalenesulfonate.
[0042] In the present specification, the "base-addition
salt" or the "salt with a base" (base-addition salt,
etc.) includes, for example, a metal salt, an inorganic
amine salt, an organic amine salt, and an amino acid
salt. The metal salt may be, for example, an alkali
metal salt such as sodium salt, potassium salt, or
lithium salt; an alkaline earth metal salt such as
calcium salt or magnesium salt; aluminum salt; iron salt;
zinc salt; copper salt; nickel salt; or cobalt salt. The
inorganic amine salt may be, for example, ammonium salt.
The organic amine salt may be, for example, morpholine
salt, glucosamine salt, ethylenediamine salt, guanidine
salt, diethylamine salt, triethylamine salt,
dicyclohexylamine salt, diethanolamine salt, piperazine
salt, or tetramethylammonium salt. Examples of the amino
acid salt include glycine salt, lysine salt, arginine
salt, ornithine salt, glutamate, and aspartate.
[0043] In the present specification, the "disease related
to integrin av" is a disease involving integrin axv and
is not particularly limited as long as its symptom is
alleviated, improved, suppressed, or prevented by
inhibiting integrin av. Integrins are heterodimers
composed of a and $ chains. The integrin av is an a
chain. Integrins comprising integrin av include
integrins axvpl, axvP3, axvP5, axvf6 and av8. The "disease
221102_G2572WO English Translation (Final) clean copy
related to integrin av" may include a disease related to
integrin avvpl, axvP3, axvP5, axvf6 or av8 because these
integrins comprise integrin av. The "disease related to
integrin av" includes, for example, a "disease related to
integrin av$l" or a "disease related to integrin av$6".
Examples of the disease related to integrin av
include inflammatory disease, fibrosis, cancer,
neurodegenerative disease, eye disease, osteoporosis, and
osteogenesis imperfecta. The inhibition of integrin av
includes decrease in the degree of signals mediated by
integrin av, as compared with a control, or decrease in
the ability of integrin av to bind to a ligand, as
compared with a control.
[0044] In the present specification, the "inflammatory
disease" refers to a disease, disorder, or a condition
related to inflammation. Examples thereof include, but
are not limited to, arthritis, rheumatoid arthritis,
Crohn disease, irritable bowel disease, inflammatory
bowel disease(IBD), systemic erythematosus, gum disease,
psoriasis, acute hepatitis, chronic hepatitis, Alport's
syndrome, inflammation caused by diabetes mellitus, joint
arterial inflammation, large-cell arterial inflammation,
Kawasaki disease, Takayasu arteritis, Churg-Strauss
syndrome and Henoch-Schonlein purpura.
[0045] In the present specification, examples of the
"fibrosis" include, but are not particularly limited to,
fibroses in the lung, the liver, the kidney, the heart,
221102_G2572W English Translation (Final) clean copy
vascular vessels, the skin, the pancreas, the bone
marrow, and other organs or tissues. In the present
invention, the fibrosis is preferably pulmonary fibrosis,
hepatic fibrosis, renal fibrosis, cardiac fibrosis,
vascular fibrosis, dermatofibrosis, pancreatic fibrosis,
myelofibrosis or skeletal myofibrosis, more preferably
pulmonary fibrosis, hepatic fibrosis, renal fibrosis or
cardiac fibrosis.
The pulmonary fibrosis includes idiopathic pulmonary
fibrosis (IPF), non-specific interstitial pneumonia
(NSIP), cryptogenic organizing pneumonia (COP), acute
interstitial pneumonia (AIP), interstitial lung disease,
peribronchiolar fibrosis, peripheral airway disease
(e.g., bronchiolitis obliterans), pulmonary emphysema,
acute respiratory distress syndrome (ARDS), acute lung
injury (ALI), postinfectious pulmonary fibrosis, drug
induced pulmonary fibrosis, airway fibrosis, diffuse
alveolar damage, collagen-vascular disease-related
pulmonary fibrosis, silicosis, radiation-induced
fibrosis, bleomycin-induced fibrosis, influenza-induced
fibrosis, asbestos-induced pulmonary fibrosis,
desquamative interstitial pneumonia and chronic
hypersensitivity pneumonitis.
The hepatic fibrosis includes primary biliary
cholangitis (PBC), primary sclerosing cholangitis (PSC),
infectious hepatic fibrosis (caused by HCV or a parasite
such as blood fluke), alcoholic steatohepatitis (ASH),
221102_G2572WO English Translation (Final) clean copy
non-alcoholic fatty liver disease (NAFLD), non-alcoholic
steatohepatitis (NASH), liver cirrhosis caused by every
etiology, congenital hepatic fibrosis, Sjogren's
syndrome, sarcoidosis, Wilson's disease, Gaucher's
disease, hemochromatosis, and al-antitrypsin deficiency.
The renal fibrosis includes chronic kidney disease
(CKD), glomerulonephritis (GN) of every etiology,
mesangial proliferative GN, immune GN, crescentic GN,
tubulointerstitial injury, renal interstitial fibrosis,
renal fibrosis brought about by complications of drug
exposure (including the cyclosporine treatment of
transplantation recipients), nephrogenic systemic
fibrosis, HIV-related nephropathy, graft nephropathy,
diabetic kidney disease (diabetic nephropathy), focal
segmental glomerulosclerosis (FSGS), idiopathic
retroperitoneal fibrosis, hypertensive nephrosclerosis
and IgA nephropathy.
The cardiac fibrosis and the vascular fibrosis
include cardiac fibrosis associated with myocardial
infarction, vascular injury-induced fibrosis, cardiac
stenosis, aortostenosis, atherosclerosis, thrombosis,
arteriosclerosis, myocardial infarction, angina pectoris,
cardiac valvular disease, renal arteriosclerosis,
congestive heart failure, dilated cardiomyopathy,
hypertrophic cardiomyopathy, restrictive cardiomyopathy,
myocarditis, vascular stenosis cardiac fibrosis, post
infarction cardiac fibrosis, left ventricular
221102_G2572WO English Translation (Final) clean copy
hypertrophy, venous occlusion, restenosis, restenosis
after angiogenesis, arteriovenous transplantation
failure, hypertension, hypertensive heart disease,
cardiomegaly, heart failure, aortic disease, hereditary
hemorrhagic telangiectasia (HHT), arterial tortuosity
syndrome, Raynaud syndrome and hypertriglyceridemia.
The dermatofibrosis includes generalized
scleroderma, polymyositis, systemic lupus erythematosus,
dermatomyositis, Dupuytren's contracture, postoperative
adhesion, fibrosis caused by surgical incision or
mechanical trauma, and excessive or hypertrophic scarring
or keloid formation in the dermis during wound healing
brought about from trauma or surgical wound.
The pancreatic fibrosis includes cystic fibrosis
(CF), chronic pancreatitis, and autoimmune pancreatitis.
The myelofibrosis includes primary myelofibrosis,
idiopathic myelofibrosis, secondary myelofibrosis, and
myeloproliferative tumor.
The fibrosis in other organs or tissues includes
skeletal myofibrosis, intestinal fibrosis, fibrosis after
organ transplantation, induratio penis plastica (Peyronie
disease), bladder fibrosis, Camurati-Engelmann disease,
Loeys-Dietz syndrome, Marfan syndrome, premature ovarian
failure, preeclampsia, mediastinal fibrosis, and
intestinal stenosis.
[0046] In the present specification, the "cancer"
includes tumor angiogenesis, carcinoma, sarcoma,
221102_G2572WO English Translation (Final) clean copy
lymphoma, leukemia, melanoma, mesothelioma, multiple
myeloma, seminoma, and tumor metastasis. Examples of the
tumor site that may be treated according to the present
invention include the bladder, blood, bone, the brain,
the breast, the central nervous system, the uterine
cervix, the colon, the endometrium, the esophagus, the
gallbladder, the genital organ, the genitourinary tract,
the head, the kidney, the larynx, the liver, the lung,
muscular tissues, the neck, oral or nasal mucosa, the
ovary, the pancreas, the prostate, the skin, the spleen,
the small intestine, the large intestine, the stomach,
the testis, and the thyroid gland.
[0047] In the present specification, the
"neurodegenerative disease" includes peripheral nerve
disorder, Alzheimer's disease, Parkinson's disease, and
multiple sclerosis.
[0048] In the present specification, the "eye disease"
includes age-related macular degeneration (AMD),
glaucoma, ocular fibrosis, retinal or corneal scarring,
diabetic retinopathy, proliferative vitreoretinopathy
(PVR), vitreoretinopathy of arbitrary etiology, fibrosis
related to ophthalmic surgery, retinal reattachment
surgery, cataract extraction, or an arbitrary type of
drainage treatment, scarring in the cornea and the
conjunctiva, corneal endothelial fibrosis, alkaline burn
(e.g., alkaline burn in the cornea), capsulolenticular
fibrosis after cataract operation, excessive scarring in
221102_G2572WO English Translation (Final) clean copy
tissues surrounding extraocular muscle in strabismus
operation, anterior subcapsular cataract and posterior
capsule opacification, anterior eye fibrosis, corneal
stromal fibrosis, fibrosis related to corneal clouding,
trabecular meshwork fibrosis, fibrosis related to
glaucoma, posterior eye fibrosis, fibrovascular scarring,
fibrosis in the retinal blood vessel or choroid blood
vessel of the eye, retinal fibrosis, epiretinal fibrosis,
retinal gliosis, subretinal fibrosis, fibrosis related to
retinal operation and glaucoma operation, retinal
traction detachment related to tissue contraction in
diabetic retinopathy, scarring of cicatricial pemphigoid
glaucoma filtration surgery, retinopathy of prematurity
(ROP) and familial exudative vitreoretinopathy (FEVR).
[0049] In the present specification, the "subject" is a
human, a monkey, a bovine, a horse, sheep, a goat, a
rabbit, a dog, a cat, a mouse, a rat, a guinea pig or a
transgenic species thereof. In the present
specification, the subject is an individual in need of
the treatment of the disease.
[0050] In the present specification, the "treatment"
includes at least one of prevention, alleviation and
remedy and also encompasses reduction of a symptom of the
disease, suppression of the progression of a symptom of
the disease, removal of a symptom of the disease,
improvement in the prognosis of the disease, prevention
of the recurrence of the disease, and prevention of the
221102_G2572WO English Translation (Final) clean copy
disease. In one aspect of the present invention, the
treatment is remedy.
[0051] <Compound of present invention>
In one embodiment, the present invention relates to
a compound represented by the following general formula
(I) or (II) or a pharmaceutically acceptable salt
thereof:
Y Y
HN N O HN N O HN N N COR HN N CO2 R 0/ OA 0O A R 1x/ R N-NH HN-N
(I) (II) wherein
A is a C6-C10 aryl group or a heteroaryl group,
wherein at least one hydrogen atom of the aryl group or
the heteroaryl group is optionally replaced with a
substituent selected from the group consisting of a
halogen atom, a hydroxy group, a C1-C6 alkyl group, a C1
C6 haloalkyl group, a C2-C6 alkenyl group, a C2-C6 alkynyl
group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a
C3-C6 cycloalkyl group, a C3-C6 cycloalkenyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, a heteroaryl
group optionally substituted by a C1-C6 alkyl group, a
cyano group, a carboxyl group, a carbamoyl group, a C1-C6
alkoxycarbonyl group, a C1-C6 alkylsulfanyl group, and a
C1-C6 alkylsulfonyl group,
221102_G2572WO English Translation (Final) clean copy
R is a hydrogen atom or a C1-C6 alkyl group,
R' is a hydrogen atom or a halogen atom, and
Y is a hydrogen atom, a fluorine atom or a hydroxy
group.
In the present specification, the compound
represented by the general formula (I) is also simply
referred to as a "compound (I)" and the compound
represented by the general formula (II) is also simply
referred to as a "compound (II)".
[0052] In one embodiment of the present invention, A in
the compound (I) or (II) is preferably a phenyl group.
At least one hydrogen atom of the phenyl group that
may be selected as A is optionally replaced with a
substituent selected from the group consisting of a
halogen atom, a hydroxy group, a C1-C6 alkyl group, a C1
C6 haloalkyl group, a C2-C6 alkenyl group, a C2-C6 alkynyl
group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a
C3-Cs6 cycloalkyl group, a C3-Cs6 cycloalkenyl group, a C3-Cs6
cycloalkoxy group, a heterocyclyl group, a heteroaryl
group optionally substituted by a C1-C6 alkyl group, a
cyano group, a carboxyl group, a carbamoyl group, a C1-C6
alkoxycarbonyl group, a C1-C6 alkylsulfanyl group, and a
C1-C6 alkylsulfonyl group.
[0053] At least one hydrogen atom of the phenyl group
that may be selected as A is optionally replaced with a
substituent selected from the group consisting of a
halogen atom, a hydroxy group, a C1-C6 alkyl group, a C1
221102_G2572WO English Translation (Final) clean copy
C6 haloalkyl group, a C2-C6 alkenyl group, a C1-C alkoxy
group, a C1-C6 haloalkoxy group, a C3-C6 cycloalkyl group,
a C3-C6 cycloalkenyl group, a C3-C6 cycloalkoxy group, a
heterocyclyl group, a heteroaryl group optionally
substituted by a C1-C alkyl group, a cyano group, a
carboxyl group, a carbamoyl group, a C1-C6 alkoxycarbonyl
group, a C1-C6 alkylsulfanyl group, and a C1-C6
alkylsulfonyl group.
[0054] In another embodiment of the present invention, at
least one hydrogen atom of the phenyl group that may be
selected as A is optionally replaced with a substituent
selected from the group consisting of a halogen atom, a
C1-C6 alkyl group, a C1-C6 haloalkyl group, a C1-C6 alkoxy
group, a C1-C6 haloalkoxy group, a C3-C6 cycloalkyl group,
a C3-C6 cycloalkoxy group, a heterocyclyl group, and a
heteroaryl group optionally substituted by a C1-C6 alkyl
group.
[0055] In an alternative embodiment of the present
invention, A in the compound (I) or (II) may be a group
represented by the following formula (i):
R6 R2
R5 RRR3
0)
[0056] In the formula (i), R 2 is a hydrogen atom or a
halogen atom.
221102_G2572WO English Translation (Final) clean copy
In another embodiment, R 2 is a hydrogen atom, a
fluorine atom or a chlorine atom.
In an alternative embodiment, R 2 is a hydrogen atom.
[0057] R3 is a hydrogen atom, a halogen atom, a C1-C6
alkyl group, a C1-C6 haloalkyl group, a C1-C6 alkoxy
group, a C1-C6 haloalkoxy group, a C3-Cs6 cycloalkyl group,
a C3-C6 cycloalkoxy group, a heterocyclyl group, or a
heteroaryl group optionally substituted by a C1-C6 alkyl
group.
In another embodiment, R 3 is a hydrogen atom, a
halogen atom, a C1-C6 haloalkyl group, a C1-C6 haloalkoxy
group, a C3-C6 cycloalkyl group, a C3-C6 cycloalkoxy
group, a heterocyclyl group, or a heteroaryl group
optionally substituted by a C1-C4 alkyl group.
In an alternative embodiment, R 3 is a hydrogen atom,
a halogen atom, a C1-C4 haloalkyl group, a C1-C4
haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group.
In an alternative embodiment, R 3 is a hydrogen atom,
a halogen atom, a difluoromethyl group, a trifluoromethyl
group, a difluoromethoxy group, a trifluoromethoxy group,
a cyclopropyl group, a cyclopropoxy group, a morpholinyl
group, or a pyrazolinyl group.
In an alternative embodiment, R 3 is a hydrogen atom,
a halogen atom, a C1-C6 alkyl group, a C1-C6 haloalkyl
group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, or
221102_G2572WO English Translation (Final) clean copy
a heteroaryl group optionally substituted by a C1-C6
alkyl group.
In an alternative embodiment, R 3 is a hydrogen atom,
a halogen atom, a C1-C4 alkyl group, a C1-C4 haloalkyl
group, a C1-C4 alkoxy group, a C1-C4 haloalkoxy group, or
a heteroaryl group optionally substituted by a C1-C4
alkyl group.
In an alternative embodiment, R 3 is a halogen atom,
a methyl group, a difluoromethyl group, a trifluoromethyl
group, a methoxy group, a difluoromethoxy group, a
trifluoromethoxy group, or a pyrazolinyl group optionally
substituted by a methyl group.
In an alternative embodiment, R 3 is a halogen atom,
a trifluoromethyl group, a difluoromethoxy group, a
trifluoromethoxy group, or a pyrazolinyl group.
[0058] R4 is a hydrogen atom, a halogen atom, a C1-C6
alkyl group, a C1-C6 haloalkyl group, a C1-C6 alkoxy
group, or a C1-C6 haloalkoxy group.
In another embodiment, R 4 is a hydrogen atom, a
halogen atom, a C1-C4 alkyl group, a C1-C4 haloalkyl
group, a C1-C4 alkoxy group, or a C1-C4 haloalkoxy group.
In an alternative embodiment, R 4 is a hydrogen atom,
a halogen atom, a methyl group, a trifluoromethyl group,
a methoxy group, or a difluoromethoxy group.
In an alternative embodiment, R 4 is a hydrogen atom
or a halogen atom.
221102_G2572WO English Translation (Final) clean copy
In an alternative embodiment, R 4 is a hydrogen atom
or a fluorine atom.
[0059] R5 is a hydrogen atom, a halogen atom, a C1-C6
alkyl group, a C1-C6 haloalkyl group, a C1-C6 alkoxy
group, a C1-C6 haloalkoxy group, or a heteroaryl group
optionally substituted by a C1-C6 alkyl group.
In another embodiment, R 5 is a hydrogen atom, a
halogen atom, a C1-C4 alkyl group, a C1-C4 haloalkyl
group, a C1-C4 alkoxy group, a C1-C4 haloalkoxy group, or
a pyrazolinyl group.
In an alternative embodiment, R5 is a hydrogen atom,
a halogen atom, a methyl group, a trifluoromethyl group,
a methoxy group, a difluoromethoxy group, or a
pyrazolinyl group.
In an alternative embodiment, R5 is a hydrogen atom,
a halogen atom, a C1-C6 alkyl group, a C1-C6 haloalkyl
group, a C1-C6 alkoxy group, or a C1-C6 haloalkoxy group.
In an alternative embodiment, R5 is a hydrogen atom,
a halogen atom, a C1-C4 alkyl group, a C1-C4 haloalkyl
group, a C1-C4 alkoxy group, or a C1-C4 haloalkoxy group.
In an alternative embodiment, R5 is a hydrogen atom,
a halogen atom, a methyl group, a difluoromethyl group, a
trifluoromethyl group, a methoxy group, a difluoromethoxy
group, or a trifluoromethoxy group.
In an alternative embodiment, R5 is a halogen atom,
a methyl group, a trifluoromethyl group, a
difluoromethoxy group, or a trifluoromethoxy group.
221102_G2572WO English Translation (Final) clean copy
[0060] R6 is a hydrogen atom or a halogen atom.
In another embodiment, R 6 is a hydrogen atom, a
fluorine atom or a chlorine atom.
In an alternative embodiment, R 6 is a hydrogen atom
or a fluorine atom.
[0061] In an alternative embodiment, for example, in the
formula (i),
R 2 may be a hydrogen atom,
R 3 may be a hydrogen atom, a halogen atom, a C1-C6
alkyl group, a C1-C6 haloalkyl group, a C1-C6 alkoxy
group, a C1-C6 haloalkoxy group, or a heteroaryl group
optionally substituted by a C1-C6 alkyl group,
R 4 may be a hydrogen atom or a halogen atom,
R5 may be a hydrogen atom, a halogen atom, a C1-C6
alkyl group, a C1-C6 haloalkyl group, a C1-C6 alkoxy
group, or a C1-C6 haloalkoxy group, and
R 6 may be a hydrogen atom or a halogen atom.
[0062] In an alternative embodiment, in the formula (i),
R 2 may be a hydrogen atom,
R 3 may be a hydrogen atom, a halogen atom, a C1-C6
haloalkyl group, a C1-C6 haloalkoxy group, a C3-C6
cycloalkyl group, a C3-C6 cycloalkoxy group, a
heterocyclyl group, or a heteroaryl group optionally
substituted by a C1-C4 alkyl group,
R 4 may be a hydrogen atom or a halogen atom,
221102_G2572WO English Translation (Final) clean copy
R 5 may be a hydrogen atom, a halogen atom, a C1-C6
alkyl group, a C1-C6 haloalkyl group, a C1-C6 alkoxy
group, or a C1-C6 haloalkoxy group, and
R 6 may be a hydrogen atom or a halogen atom.
[0063] In an alternative embodiment of the present
invention, A in the compound (I) or (II) may be a group
represented by any of the following formulas (i)-a to
(i)-g:
RaR R~ 6 12 Ral Ra2 R a Ra R'311 Ra Ra Ralo R
(i)-a (i)-b (i)-c (i)-d (i)-e (i)-f (i-g
wherein * represents a binding position, and Rai to Ra13 are each independently a substituent
selected from the group consisting of a halogen atom, a
hydroxy group, a C1-C6 alkyl group, a C1-C6 haloalkyl
group, a C2-C6 alkenyl group, a C2-C6 alkynyl group, a C1
C6 alkoxy group, a C1-C6 haloalkoxy group, a C3-C6
cycloalkyl group, a C3-C6 cycloalkenyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, a heteroaryl
group optionally substituted by a C1-C6 alkyl group, a
cyano group, a carboxyl group, a carbamoyl group, a C1-C6
alkoxycarbonyl group, a C1-C6 alkylsulfanyl group, and a
C1-C6 alkylsulfonyl group. Rai
[0064] In the embodiment, to Ra 1 3 may each
independently be a substituent selected from the group
consisting of a halogen atom, a C1-C6 alkyl group, a C1-C6
221102_G2572WO English Translation (Final) clean copy
haloalkyl group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy
group, a C3-C6 cycloalkyl group, a C3-C6 cycloalkoxy
group, a heterocyclyl group, and a heteroaryl group
optionally substituted by a C1-C6 alkyl group.
[0065] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-a,
one of Rai and Ra 2 may be a halogen atom, a C1-C6
haloalkyl group, or a C1-C6 haloalkoxy group, and
the other moiety may be a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
C1-C6 haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group optionally substituted by a C1-C6 alkyl group.
[0066] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-a, Rai one of and Ra 2 may be a halogen atom, a
trifluoromethyl group, a difluoromethoxy group, or a
trifluoromethoxy group, and
the other moiety may be a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
C1-C6 haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group optionally substituted by a C1-C6 alkyl group.
221102_G2572WO English Translation (Final) clean copy
[0067] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-a,
one of Rai and Ra 2 may be a halogen atom, a
difluoromethyl group, a trifluoromethyl group, a
difluoromethoxy group, or a trifluoromethoxy group, and
the other moiety may be a halogen atom, a methyl
group, a difluoromethyl group, a trifluoromethyl group, a
methoxy group, a difluoromethoxy group, a
trifluoromethoxy group, a cyclopropyl group, a
cyclopropoxy group, a morpholinyl group, or a pyrazolinyl
group.
[0068] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-b,
Ra3 may be a halogen atom, and
Ra 4 may be a halogen atom, a C1-C6 alkyl group, a C1
C6 haloalkyl group, a C1-C6 alkoxy group, a C1-C6
haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group optionally substituted by a C1-C6 alkyl group.
[0069] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-b,
Ra3 may be a halogen atom, and
Ra 4 may be a halogen atom, a C1-C6 haloalkyl group,
or a C1-C6 haloalkoxy group.
221102_G2572WO English Translation (Final) clean copy
[0070] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-b,
Ra3 may be a halogen atom, and
Ra 4 may be a C1-C6 haloalkyl group.
[0071] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-c,
Ra5 may be a halogen atom, and
Ra6 may be a halogen atom, a C1-C6 alkyl group, a C1
C6 haloalkyl group, a C1-C6 alkoxy group, a C1-C6
haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group optionally substituted by a C1-C6 alkyl group.
[0072] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-c,
Ra5 may be a halogen atom, and
Ra6 may be a halogen atom, a C1-C6 haloalkyl group,
or a C1-C6 haloalkoxy group.
[0073] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-c,
Ra5 may be a halogen atom, and
Ra6 may be a halogen atom, a trifluoromethyl group,
or a trifluoromethoxy group.
221102_G2572WO English Translation (Final) clean copy
[0074] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-d,
one of Ra7 and Ra8 may be a halogen atom, a C1-C6
haloalkyl group, or a C1-C6 haloalkoxy group, and
the other moiety may be a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
C1-C haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group optionally substituted by a C1-C6 alkyl group.
[0075] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-d,
one of Ra7 and Ra8 may be a halogen atom, a C1-C6
haloalkyl group, or a C1-C6 haloalkoxy group, and
the other moiety may be a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, or a
C1-C6 haloalkoxy group.
[0076] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-d,
one of Ra7 and Ra8 may be a halogen atom, a
trifluoromethyl group, or a trifluoromethoxy group, and
the other moiety may be a halogen atom, a methyl
group, a difluoromethyl group, a trifluoromethyl group, a
methoxy group, a difluoromethoxy group, or a
trifluoromethoxy group.
221102_G2572WO English Translation (Final) clean copy
[0077] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-e, Raio at least one of Rag, and Ral may be a halogen
atom, a C1-C6 haloalkyl group, or a C1-C6 haloalkoxy
group, and
the remaining moieties may each individually be a
halogen atom, a C1-C6 alkyl group, a C1-C6 haloalkyl
group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a
C3-C6 cycloalkyl group, a C3-C6 cycloalkoxy group, a
heterocyclyl group, or a heteroaryl group optionally
substituted by a C1-C6 alkyl group.
[0078] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-e, Raio Ra9, and Ra1 1 may each individually be a halogen
atom, a C1-C6 haloalkyl group, or a C1-C6 haloalkoxy
group.
[0079] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-f,
Ra 1 2 may be a halogen atom, a C1-C6 haloalkyl group, a
C1-C6 haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group optionally substituted by a C1-C6 alkyl group.
221102_G2572WO English Translation (Final) clean copy
[0080] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-f,
Ra1 2 may be a halogen atom, a difluoromethyl group, a
trifluoromethyl group, a difluoromethoxy group, a
trifluoromethoxy group, a cyclopropyl group, a
cyclopropoxy group, a morpholinyl group, or a pyrazolinyl
group optionally substituted by a methyl group.
[0081] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-g,
Ra1 3 may be a halogen atom, a C1-C6 haloalkyl group, a
C1-C6 haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group optionally substituted by a C1-C6 alkyl group.
[0082] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-f,
Rai 3 may be a halogen atom, a difluoromethyl group, a
trifluoromethyl group, a difluoromethoxy group, a
trifluoromethoxy group, a cyclopropyl group, a
cyclopropoxy group, a morpholinyl group, or a pyrazolinyl
group optionally substituted by a methyl group.
[0083] In an alternative embodiment of the present
invention, when A in the compound (I) or (II) is a group
represented by the formula (i)-f,
221102_G2572WO English Translation (Final) clean copy
Rai3 may be a C1-C6 haloalkyl group or a C1-C6
haloalkoxy group.
Alternatively, Rais may be a difluoromethyl group, a
trifluoromethyl group, a difluoromethoxy group, or a
trifluoromethoxy group.
[0084] In one embodiment, R in the compound (I) or (II)
is a hydrogen atom or a C1-C alkyl group.
In another embodiment, R in the compound (I) or (II)
is a hydrogen atom or a C1-C4 alkyl group.
In an alternative embodiment, R in the compound (I)
or (II) is a hydrogen atom or a methyl group.
In an alternative embodiment, R in the compound (I)
or (II) is a hydrogen atom.
[0085] In one embodiment, R' in the compound (I) or (II)
is a hydrogen atom or a halogen atom.
In another embodiment, R' in the compound (I) or
(II) is a hydrogen atom, a fluorine atom or a chlorine
atom.
In an alternative embodiment, R' in the compound (I)
or (II) is a hydrogen atom.
[0086] In one embodiment, Y in the compound (I) or (II)
is a hydrogen atom, a fluorine atom or a hydroxy group.
In an alternative embodiment, Y in the compound (I)
or (II) is a fluorine atom.
[0087] In an alternative embodiment, the present
invention provides a compound selected from the following
group or a pharmaceutically acceptable salt thereof:
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-(trifluoromethyl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
iodo-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
methyl-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
methoxy-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
fluoro-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(3,5-bis(trifluoromethyl)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(1H-pyrazol-1-yl)-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(2
fluoro-3-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-chloro-3-(trifluoromethyl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(2
fluoro-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
fluoro-3-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
methyl-3-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(3-chloro-4-fluoro-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-4-methoxyphenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3,5-dichlorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-chlorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3,5-dibromophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-iodophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-chloro-5-iodophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(5-chloro-2-fluorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(5-bromo-2-fluorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(difluoromethoxy)-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3-chloro-5-(difluoromethoxy)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-(difluoromethoxy)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(difluoromethoxy)-4-fluorophenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(5-(difluoromethoxy)-2-fluorophenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(difluoromethoxy)-5-(1H-pyrazol-1
yl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3,5-bis(difluoromethoxy)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-chloro-5-(trifluoromethoxy)phenyl)-3-(2
(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-(trifluoromethoxy)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(2
fluoro-5-(trifluoromethoxy)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
fluoro-3-(trifluoromethoxy)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
(trifluoromethoxy)phenyl)propanoic acid,
(3S)-3-(3-(difluoromethoxy)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(difluoromethyl)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-cyclopropylphenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-cyclopropoxyphenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-chlorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3-bromophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-(3,5-dimethyl-1H-pyrazol-1-yl)phenyl)-3
(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)
1H-indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
morpholinophenyl)propanoic acid,
(3S)-3-(4-(difluoromethoxy)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(3
fluoro-4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2
yl)amino)-1H-indazole-6-carboxamido)acetamido)propanoic
acid,
(3S)-3-(2-(3-chloro-4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(3-chloro-5
(trifluoromethyl)phenyl)propanoic acid,
221102_G2572WO English Translation (Final) clean copy
(3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
((5-hydroxy-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid,
(S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
((1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid,
(3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(6
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-4-carboxamido)acetamido)propanoic acid,
(3S)-3-(3-bromo-5-(trifluoromethyl)phenyl)-3-(2-(6
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-4-carboxamido)acetamido)propanoic acid,
(3S)-3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-4-carboxamido)acetamido)-3-(3
iodo-5-(trifluoromethyl)phenyl)propanoic acid,
(3S)-3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-4-carboxamido)acetamido)-3-(4
fluoro-3-(trifluoromethyl)phenyl)propanoic acid, and
(3S)-3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-4-carboxamido)acetamido)-3-(3
(trifluoromethyl)phenyl)propanoic acid.
[0088] The compound represented by the general formula
(I) or (II) of the present invention or a
pharmaceutically acceptable salt thereof can form a
hydrate or a solvate, and each individual or a mixture
thereof is encompassed by the present invention.
221102_G2572WO English Translation (Final) clean copy
[0089] When the compound represented by the general
formula (I) or (II) of the present invention or a
pharmaceutically acceptable salt thereof has one or more
asymmetric centers, carbon-carbon double bonds, axial
chirality, or the like, optical isomers (including
enantiomers and diastereomers), geometrical isomers,
tautomers, and rotational isomers may exist. These
isomers and mixtures thereof are represented by a single
formula such as the formula (I) or (II). The present
invention encompasses each of these isomers and mixtures
(including racemates) thereof at arbitrary ratios.
[0090] In an embodiment, the compound (I) of the present
invention has both the following absolute configurations:
Y Y HN N HN N
HN HCOR and HN-f N COR
R10 A R10 A N-NH N-NH
[0091] In an embodiment, the compound (I) of the present
invention has the following absolute configuration:
Y
HN N N HN N N CO 2R
N-NH
[0092] In an embodiment, the compound (I) of the present
invention has the following absolute configuration:
221102_G2572WO English Translation (Final) clean copy
Y
HN NN HN N ' CO 2 R
/ O A R1 N-NH
[0093] Likewise, in an embodiment, the compound (II) of
the present invention has both the following absolute
configurations:
Y Y HN N H HNN O H
HN N R CO2R and HN N N COR H 0 A I H 0 A
HN-N HN-N
[0094] In an embodiment, the compound (II) of the present
invention has the following absolute configuration:
Y
HN N O H HN N - I H 0 A
HN-N
[0095] In an embodiment, the compound (II) of the present
invention has the following absolute configuration:
Y
HN NNO H HN O NNCO A 2R HN H N--j A HN-N
221102_G2572WO English Translation (Final) clean copy
[0096] A racemate can be resolved into optical
enantiomers by a method known in the art, for example,
separation of a diastereoisomeric salt thereof with an
optically active acid, and release of an optically active
amine compound by treatment with a base. Another method
for resolving a racemic compound into optical enantiomers
is based on chromatography of optically active matrix.
The compound of the present invention may also be
resolved by the formation of a diastereoisomeric
derivative. A further method for resolving optical
isomers known to those skilled in the art, such as J.
Jaques, A. Collet and S. Wilen et al., "Enantiomers,
Racemates, and Resolutions", John Wiley and Sons, New
York (1981) can be used. An optically active compound
can also be produced from an optically active starting
material.
[0097] The compound represented by the general formula
(I) or (II) of the present invention can form an acid
addition salt, etc., and such an acid-addition salt, etc.
is encompassed by the pharmaceutically acceptable salt of
the present invention.
The compound represented by the general formula (I)
or (II) of the present invention can form an acid
addition salt with an acid at an arbitrary ratio, and
each individual (e.g., monohydrochloride or
dihydrochloride) or a mixture thereof is encompassed by
the present invention. When R in the general formula (I)
221102_G2572WO English Translation (Final) clean copy
or (II) is, for example, a hydrogen atom, such a compound
can form a base-addition salt, etc., and such a base
addition salt, etc. is also encompassed by the
pharmaceutically acceptable salt of the present
invention.
[0098] The compound represented by the general formula
(I) or (II) of the present invention, when having a group
capable of forming an ester group, such as a hydroxy
group or a carboxy group, can be converted to a
pharmaceutically acceptable ester, and such a
pharmaceutically acceptable ester is encompassed by the
present invention. The pharmaceutically acceptable ester
of the compound represented by the general formula (I) or
(II) is capable of serving as a prodrug of the compound
represented by the general formula (I) or (II) and can be
hydrolyzed in a metabolic process (e.g., hydrolysis),
when administered in vivo to a subject, to form the
compound represented by the general formula (I) or (II).
[0099] The compound represented by the general formula
(I) or (II) of the present invention or a
pharmaceutically acceptable salt thereof can form an
isotope compound in which one or more atoms constituting
the compound or the salt are replaced with isotope atoms
at a non-natural ratio. The isotope atom may be
radioactive or nonradioactive and includes, for example,
heavy hydrogen ( 2 H; D), tritium ( 3 H; T), carbon-14 (14C),
and iodine-125 (125I). The radioactive or nonradioactive
221102_G2572WO English Translation (Final) clean copy
isotope compound may be used as a medicament for the
remedy or prevention of a disease, a reagent for research
(e.g., a reagent for assay), a diagnostic drug (e.g., a
diagnostic imaging drug), or the like. The present
invention encompasses such a radioactive or
nonradioactive isotope compound.
[0100] The compound represented by the general formula
(I) or (II) of the present invention or a
pharmaceutically acceptable salt thereof is capable of
inhibiting the binding of integrin av to its ligand.
In an alternative embodiment, the compound
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof is capable of inhibiting the activation of TGF-.
TGF- participates in the differentiation, survival,
proliferation, and the like of cells and plays an
important role in development, immunity, wound healing,
and the like and therefore, is deeply involved in
diseases such as inflammation, fibrosis and cancer. TGF
$ is known to have three types of isoforms in mammals,
and TGF-1 is known as a master regulator for tissue
repair, inflammation, and fibrosis (Seminars in Cell and
Developmental Biology, 2020, 101, 123-139). TGF- is
secreted as an inactive form (latent form) and activated
by various methods so as to exhibit physiological
activity. RGD integrin including av1 and av6 is known
to play an important role in the activation of TGF-31.
221102_G2572WO English Translation (Final) clean copy
For example, an avl integrin inhibitor (Non Patent
Literature 8: Science Translational Medicine, 2015, 7
(288), 288ra79) or an av$6 integrin inhibitor (Cancer
Research, 2008, 68 (2), 561-570) inhibits the activation
of TGF-1. Thus, the compound represented by the general
formula (I) or (II) of the present invention or a
pharmaceutically acceptable salt thereof can be useful in
the prevention, alleviation and/or remedy of diseases
such as inflammatory disease, fibrosis, cancer,
neurodegenerative disease, eye disease, osteoporosis and
osteogenesis imperfecta.
In an alternative embodiment, the compound
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof is capable of inducing the dedifferentiation of
activated human hepatic stellate cells. Stellate cells
or fibroblasts differentiate into active stellate cells
or myofibroblasts expressing a-smooth muscle actin (ax
SMA, ACTA2) through TGF-1 or the like, and enhance
collagen production, etc. For example, it has been
reported that in integrin av-knockout stellate cells, the
expression of c-SMA was suppressed and further the
production of collagen or the like was also suppressed,
because TGF-1 is not activated (Non Patent Literature
16: Nature Medicine, 2013, 19 (12), 1617-1624). Such
enhanced collagen production is seen in diseases such as
inflammation, fibrosis and cancer. Thus, the compound
221102_G2572WO English Translation (Final) clean copy
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof can be useful in the prevention, alleviation
and/or remedy of diseases such as inflammatory disease,
fibrosis, cancer, neurodegenerative disease, eye disease,
osteoporosis and osteogenesis imperfecta.
In an alternative embodiment, the compound
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof is capable of suppressing increase in
hydroxyproline (HYP) level which is an index for fibrosis
in non-alcoholic steatohepatitis (NASH) model animals.
Thus, the compound represented by the general formula (I)
or (II) of the present invention or a pharmaceutically
acceptable salt thereof can be useful in the prevention,
alleviation and/or remedy of non-alcoholic
steatohepatitis (NASH).
In an alternative embodiment, the compound
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof has excellent pharmacokinetic characteristics.
The compound represented by the general formula (I) or
(II) of the present invention or a pharmaceutically
acceptable salt thereof is capable of exhibiting
excellent transcellular permeability, for example, in a
membrane permeability test using Caco2 cells. The
transcellular permeability in a membrane permeability
221102_G2572WO English Translation (Final) clean copy
test using Caco2 cells reportedly correlates with the
rate of intestinal absorption.
In an alternative embodiment, the compound
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof has excellent safety. The compound represented
by the general formula (I) or (II) of the present
invention or a pharmaceutically acceptable salt thereof
is shown to form no or few reactive metabolites through
metabolism in liver microsomes, for example.
[0101] <Pharmaceutical composition of present invention>
In one embodiment, the present invention relates to
a pharmaceutical composition comprising the compound
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof.
[0102] In one embodiment, the pharmaceutical composition
of the present invention can be used for the treatment of
a disease related to integrin av.
[0103] In one embodiment, the present invention relates
to use of the compound represented by the general formula
(I) or (II) of the present invention or a
pharmaceutically acceptable salt thereof for a disease
related to integrin av.
[0104] In one embodiment, the present invention relates
to the compound represented by the general formula (I) or
(II) of the present invention or a pharmaceutically
221102_G2572WO English Translation (Final) clean copy
acceptable salt thereof for use in the treatment of a
disease related to integrin av.
[0105] In one embodiment, the present invention relates
to use of the compound represented by the general formula
(I) or (II) of the present invention or a
pharmaceutically acceptable salt thereof for producing a
medicament for the treatment of a disease related to
integrin av.
[0106] In one embodiment, the present invention relates
to a method for the treatment of a disease related to
integrin av, comprising administering the compound
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof in an amount effective for prevention,
alleviation and/or remedy to a subject.
[0107] In one embodiment of the present invention,
examples of the disease related to integrin av can
include inflammatory disease, fibrosis, cancer,
neurodegenerative disease, eye disease, osteoporosis, and
osteogenesis imperfecta.
In an alternative embodiment, the disease related to
integrin av is fibrosis.
[0108] In one embodiment, the fibrosis can be selected
from fibroses in the lung, the liver, the kidney, the
heart, vascular vessels, the skin, the pancreas, the bone
marrow, and other organs or tissues.
221102_G2572WO English Translation (Final) clean copy
In another embodiment, the fibrosis can be selected
from pulmonary fibrosis, hepatic fibrosis, renal
fibrosis, cardiac fibrosis, vascular fibrosis,
dermatofibrosis, pancreatic fibrosis, myelofibrosis and
skeletal myofibrosis.
In an alternative embodiment, the fibrosis can be
selected from pulmonary fibrosis, hepatic fibrosis, renal
fibrosis and cardiac fibrosis.
[0109] In one embodiment, the disease related to integrin
axv is idiopathic pulmonary fibrosis (IPF).
In another embodiment, the disease related to
integrin av is non-alcoholic fatty liver disease (NAFLD).
In an alternative embodiment, the disease related to
integrin av is non-alcoholic steatohepatitis (NASH).
In an alternative embodiment, the disease related to
integrin av is primary biliary cholangitis (PBC).
In an alternative embodiment, the disease related to
integrin av is primary sclerosing cholangitis (PSC).
In an alternative embodiment, the disease related to
integrin av is alcoholic steatohepatitis.
In an alternative embodiment, the disease related to
integrin av is chronic kidney disease (CKD).
In an alternative embodiment, the disease related to
integrin av is diabetic kidney disease (diabetic
nephropathy).
221102_G2572WO English Translation (Final) clean copy
In an alternative embodiment, the disease related to
integrin av is cardiac fibrosis associated with
myocardial infarction.
[0110] The subject to which the compound represented by
the general formula (I) or (II) of the present invention
or a pharmaceutically acceptable salt thereof or the
pharmaceutical composition of the present invention is
administered is a mammal. The mammal includes, for
example, a human, a monkey, a bovine, a horse, sheep, a
goat, a rabbit, a dog, a cat, a mouse, a rat, a guinea
pig or a transgenic species thereof. The compound
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof or the pharmaceutical composition of the present
invention is preferably administered to a human.
[0111] The dose, i.e., the amount effective for
prevention, alleviation and/or remedy, of the compound
represented by the general formula (I) or (II) of the
present invention or a pharmaceutically acceptable salt
thereof may vary depending on the frequency and method of
administration, the animal species, sex, age, body weight
and general condition of a recipient, the severity of the
disease, etc., and can be appropriately set by those
skilled in the art. For oral administration as to an
adult human (60 kg), for example, 0.01 to 3000 mg,
preferably 0.1 to 600 mg, is appropriate as the dose per
day of the compound of the present invention and can be
221102_G2572WO English Translation (Final) clean copy
administered once to several times a day, for example, in
one portion or two, three or four divided portions. For
parenteral administration to an adult human, for example,
0.001 to 1000 mg, preferably 0.01 to 300 mg, is
appropriate as the dose of the compound of the present
invention and can be administered once to several times a
day, for example, in one portion or two, three or four
divided portions. For parenteral administration, the
compound of the present invention can be used at a
concentration of, for example, 0.00001% (w/v) to 10%
(w/v), preferably 0.001% (w/v) to 5% (w/v).
[0112] As for a parenteral route of intravenous,
intrathecal, intramuscular or similar administration,
etc., the dosage is typically smaller than an amount that
is used for oral administration.
[0113] The compound represented by the general formula
(I) or (II) of the present invention or a
pharmaceutically acceptable salt thereof may be
administered by single administration or multiple
administration, either singly as a pure compound or in
combination with a pharmaceutically acceptable carrier.
A pharmaceutical composition formed by combining the
compound of the present invention with a pharmaceutically
acceptable carrier may be administered in various dosage
forms suitable for administration routes. The
pharmaceutical composition according to the present
invention may be formulated together with a
221102_G2572WO English Translation (Final) clean copy
pharmaceutically acceptable carrier in accordance with a
conventional approach disclosed in, for example,
Remington: The Science and Practice of Pharmacy, 19
Edition, Gennaro, Ed., Mack Publishing Co., Easton, PA,
1995.
[0114] The pharmaceutical composition of the present
invention can be administered orally or parenterally.
The parenteral route may include, for example, rectal,
transnasal, pulmonary, transdermal, intracerebral,
intraperitoneal, vaginal, subcutaneous, intramuscular,
intrathecal, intravenous and intradermal routes. The
pharmaceutical composition of the present invention may
be specified and formulated for administration through an
arbitrary appropriate route. A preferred route may be
selected depending on the general condition and age of a
recipient, the properties of a condition to receive a
treatment, and an active ingredient selected.
[0115] The pharmaceutical composition for oral
administration includes solid preparations such as
capsules, tablets, sugar-coated tablets, pills, lozenges,
powders and granules. Such solid preparations can be
prepared in a coated form, if appropriate. Nonsolid
preparations for oral administrations include solutions
(including solutions for inhalations), emulsions,
suspensions, syrups and elixirs.
[0116] The pharmaceutical composition for parenteral
administration includes, for example, injections,
221102_G2572WO English Translation (Final) clean copy
suppositories, sprays, ointments, creams, gels,
inhalants, skin patches, and implants. The injection is
particularly suitable for intravenous, intramuscular,
subcutaneous and intraperitoneal administration. Every
sterile aqueous medium used is readily available by a
standard approach known to those skilled in the art.
[0117] In the case of preparing an oral solid
preparation, a carrier, for example, an excipient, and
optionally a binder, a disintegrant, a lubricant, a
colorant, a corrigent, and the like can be added to a
principal agent, followed by a routine method to prepare
tablets, coated tablets, granules, fine granules,
powders, capsules, or the like. An oral nonsolid
preparation such as a syrup can also be appropriately
prepared.
[0118] For example, lactose, sucrose, corn starch,
saccharose, glucose, sorbitol, crystalline cellulose, or
silicon dioxide is used as the excipient. For example,
polyvinyl alcohol, ethylcellulose, methylcellulose, gum
arabic, hydroxypropylcellulose, or
hydroxypropylmethylcellulose is used as the binder. For
example, magnesium stearate, talc, or silica is used as
the lubricant. An agent that is acceptable for addition
to medicaments is used as the colorant. For example,
cacao powder, menthol, aromatic acid, mint oil, kapur, or
powdered cinnamon bark is used as the corrigent. Such
tablets or granules may be appropriately coated, if
221102_G2572WO English Translation (Final) clean copy
necessary, with sugar, gelatin, or the like as a matter
of course.
[0119] In the case of preparing an injection, a carrier,
for example, a pH adjuster, a buffer, a suspending agent,
a solubilizer, a stabilizer, a tonicity agent, or a
preservative can be added, if necessary, to a principal
agent, followed by a routine method to prepare an
intravenous injection, a subcutaneous injection, an
intramuscular injection, or an intravenous drip infusion
agent. In this respect, a freeze-dried product may be
prepared by a routine method, if necessary.
[0120] Examples of the suspending agent can include
methylcellulose, polysorbate 80, hydroxyethylcellulose,
gum arabic, powdered tragacanth, sodium
carboxymethylcellulose, and polyoxyethylene sorbitan
monolaurate.
[0121] Examples of the solubilizer can include
polyoxyethylene hydrogenated castor oil, polysorbate 80,
nicotinamide, polyoxyethylene sorbitan monolaurate,
macrogol, castor oil fatty acid ethyl ester, and
cyclodextrin.
[0122] Examples of the stabilizer can include sodium
sulfite and sodium metasulfite. Examples of the
preservative can include methyl p-hydroxybenzoate, ethyl
p-hydroxybenzoate, sorbic acid, phenol, cresol, and
chlorocresol.
[0123] <Synthesis method>
221102_G2572WO English Translation (Final) clean copy
The compound represented by the general formula (I)
or (II) of the present invention can be produced by a
method summarized in any of reaction processes 1 to 2
given below. A modification or an alteration that is
known per se to chemists in the art or can be obvious to
those skilled in the art may be used in the methods
described below.
[0124] In the following description about production
methods, A, R, R1 and Y are as defined above, unless
otherwise specified.
[0125] <Process 1>
A compound (I) or (II) wherein R is a Ci-C6 alkyl
group can be produced through step 1 of the following
process 1.
Y Y
HN N H HN N H O H2N N CO 2 R H 0O HN OH + 0 A ' H N CO2R R 1 (3) Step 1 R0 O A N-NH N-NH
(1A) (i)
Y Y
HN-.-N 0 H HN.-N o N2 HT HN N OH H2N CO2 R HN N H 0 A + A Sep 1 1; R (3) Ste1 HN-N HN-N
(2A) (II)
[0126] Step 1 of process 1 is the step of condensing a
compound (1A) or (2A) and a compound (3) in a solvent
221102_G2572WO English Translation (Final) clean copy
using a dehydration-condensation agent to produce the
compound (I) or (II) of the present invention.
The compound (1A) or (2A) can be produced in
accordance with syntheses 1 and 2 mentioned later and the
description of Examples of the present specification.
The compound (1A) or (2A) may be an acid-addition salt,
etc. Examples of the acid-addition salt, etc. include
hydrochloride, sulfate, and acetate.
[0127] The compound (3) can also be produced in
accordance with synthesis 6 mentioned later and the
description of Examples of the present specification.
The compound (3) may be an acid-addition salt, etc.
Examples of the acid-addition salt, etc. include
hydrochloride, sulfate, and acetate.
[0128] Examples of the dehydration-condensation agent for
use in step 1 of process 1 include
dicyclohexylcarbodiimide (DCC), N,N'
diisopropylcarbodiimide (DIC), 1-(3-dimethylaminopropyl)
3-ethylcarbodiimide or hydrochloride thereof (EDC, EDAC),
N,N-dicyclohexylamide, carbonyldiimidazole, 1H
benzotriazol-1-yloxytris(dimethylamino)phosphonium
hexafluorophosphate, and 0-(7-azabenzotriazol-1-yl)
1,1,3,3-tetramethyluronium hexafluorophosphate. N,N'
Diisopropylcarbodiimide or dicyclohexylcarbodiimide is
preferred.
[0129] Step 1 of process 1 may be performed, if
necessary, in the presence of an activator such as 1
221102_G2572WO English Translation (Final) clean copy
hydroxybenzotriazole (HOBt), 1-hydroxy-7-azabenzotriazole
(HOAt), N-hydroxysuccinimide (HOSu), or 4
dimethylaminopyridine.
Step 1 of process 1 may be performed, if necessary,
in the presence of a base such as N,N
diisopropylethylamine, N-methylmorpholine, or
triethylamine.
[0130] The solvent for use in step 1 of process 1 is not
particularly limited as long as the solvent dissolves the
starting materials to some extent without inhibiting the
reaction. Examples thereof include: amides such as N,N
dimethylformamide (DMF), N,N-dimethylacetamide, and N
methylpyrrolidone; halogenated hydrocarbons such as
dichloromethane (DCM); esters such as ethyl acetate;
hydrocarbons such as dichlorohexane and n-hexane;
aromatic hydrocarbons such as toluene; ethers such as
tetrahydrofuran, diethyl ether, cyclopentyl methyl ether,
1,4-dioxane, and tert-butyl methyl ether; sulfur solvents
such as dimethyl sulfoxide (DMSO); and arbitrary mixed
solvents thereof. An aprotic polar solvent is preferred,
and N,N-dimethylformamide, dimethyl sulfoxide, or a mixed
solvent prepared by mixing them is more preferred.
[0131] The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually -20°C to 100°C,
preferably 0°C to 50°C.
221102_G2572WO English Translation (Final) clean copy
[0132] The reaction time differs depending on the
reaction temperature, etc., and is usually 10 minutes to
120 hours, preferably 30 minutes to 48 hours.
[0133] <Process 2>
A compound (I') or (II') which is a compound (I) or
(II) wherein R is a hydrogen atom can be produced by
subjecting the compound (I) or (II) produced by the
process 1 to step 2 of the following process 2.
Y Y
HN N HN N Y 0 H 0 H HN N N CO2R 3 HN N CO2H RI H 02 Step 2 RI H 02
N-NH N-NH
(1) (1)
Y Y
HN -N 0 HN N TY H 0 H HN N N CO2R 3 HN R N CO2H H0 A Step 2 H 0 A
HN-N HN-N (II) (II')
[0134] Step 2 of process 2 is the step of hydrolyzing the
ester moiety of the compound (I) or (II) or an acid
addition salt, etc. thereof in a solvent, for example,
through reaction with a base to produce a compound (I')
or (II').
[0135] Examples of the base for use in step 2 of process
2 include alkali metal hydroxide such as lithium
hydroxide, sodium hydroxide, and potassium hydroxide, and
221102_G2572WO English Translation (Final) clean copy
alkaline earth metal hydroxide such as calcium hydroxide
and magnesium hydroxide. Alkali metal hydroxide is
preferred, and lithium hydroxide or sodium hydroxide is
more preferred. These bases may each be used singly or
may be used in combination of two or more thereof. A
method for adding the base may involve adding the base in
the form of a solution containing the base dissolved in a
solvent mentioned later.
[0136] The solvent for use in step 2 of process 2 is not
particularly limited as long as the solvent dissolves the
starting materials to some extent without inhibiting the
reaction. Examples thereof include: water; amides such
as N,N-dimethylformamide, N,N-dimethylacetamide, and N
methylpyrrolidone; halogenated hydrocarbons such as
dichloromethane; hydrocarbons such as dichlorohexane and
n-hexane; aromatic hydrocarbons such as toluene; ethers
such as tetrahydrofuran, diethyl ether, cyclopentyl
methyl ether, 1,4-dioxane, and tert-butyl methyl ether;
alcohols such as methanol, ethanol, n-propanol, and
isopropanol; nitriles such as acetonitrile and
propionitrile; and arbitrary mixed solvents thereof.
Water, acetonitrile, tetrahydrofuran, N,N
dimethylformamide, methanol, ethanol, or a mixed solvent
prepared by mixing two or more of them is preferred.
[0137] The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
221102_G2572WO English Translation (Final) clean copy
solvent, and the like, and is usually -20°C to 100°C,
preferably 00C to 50°C.
[0138] The reaction time differs depending on the
reaction temperature, etc., and is usually 1 minute to 48
hours, preferably 10 minutes to 30 hours.
[0139] <Synthesis 1>
The compounds (1A) and (2A) for use as starting
materials in the process 1 mentioned above can be
produced through each step of the following synthesis 1.
221102_G2572WO English Translation (Final) clean copy
O HN S O H2N ORx CH 3NCS HN ORx
R1 R Step 3 Step 4 N-Ns 1 N-N
(1A)-1 (1A)-2
1) Mel HN S 2) NH HN 2 HN -N I OH T0 Ral \ o HN OH
N-NH Step5 N-NH (1A)-3 (1A)
H2NH H H2N ORx CH 3NCS N ORx Ri S 1 ,NN Step 3 ,N-N R/R1 g/N9 4 Step 1 N-N
(2A)-1 (2A)-2
1) Mel NH 2 H H 2) H N NOH (NH2 HN-N O S R(al S HN OH HN-N StepR5 HN-N (2A)-3 (2A)
[0140] In the formulas, P1 represents a protective group
such as a tetrahydropyranyl (THP) group.
221102_G2572WO English Translation (Final) clean copy
In the formulas, Rx is a hydrocarbon group having 1
to 6 carbon atoms, specifically, preferably a C1-C6 alkyl
group.
In the following description about production
methods, P' and Rx are as defined above unless otherwise
specified.
[0141] Step 3 of synthesis 1 is the step of reacting a
compound (1A)-1 or (2A)-1 with methyl isothiocyanate in a
solvent to obtain a compound (1A)-2 or (2A)-2.
The compound (1A)-1 or (2A)-1 is known in the art or
may be produced from another compound known in the art.
The compound can be produced, for example, in accordance
with syntheses 3 and 4 mentioned later and the
description of Examples of the present specification.
[0142] The solvent for use in step 3 of synthesis 1 is
not particularly limited as long as the solvent dissolves
the starting materials to some extent without inhibiting
the reaction. Examples thereof include: amides such as
N,N-dimethylformamide, N,N-dimethylacetamide, and N
methylpyrrolidone; halogenated hydrocarbons such as
dichloromethane; esters such as ethyl acetate;
hydrocarbons such as dichlorohexane and n-hexane;
aromatic hydrocarbons such as toluene; ethers such as
tetrahydrofuran, diethyl ether, cyclopentyl methyl ether,
1,4-dioxane, and tert-butyl methyl ether; alcohols such
as methanol, ethanol, n-propanol, and isopropanol;
nitriles such as acetonitrile and propionitrile; and
221102_G2572WO English Translation (Final) clean copy
arbitrary mixed solvents thereof. N,N-Dimethylformamide,
N-methylpyrrolidone, or methanol is preferred.
Step 3 of synthesis 1 may be performed, if
necessary, in the presence of acetic acid.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually 0°C to 150°C,
preferably 200C to 120°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 72 hours,
preferably 30 minutes to 48 hours.
[0143] Step 4 of synthesis 1 is the step of eliminating
the protective group P' and the substituent Rx in the
compound (1A)-2 or (2A)-2 in a solvent to obtain a
compound (1A)-3 or (2A)-3.
In step 4 of synthesis 1, the protective group P'
may be eliminated first, or the substituent Rx may be
eliminated first.
[0144] When the protective group P' is, for example, a
tetrahydropyranyl group, this group can be eliminated by
preparing an acidic solution by the addition of an acid
to a solution containing the compound (1A)-2 or (2A)-2.
Examples of the acid for use in the elimination of
the protective group P' in step 4 of synthesis 1 include,
but are not particularly limited to, hydrochloric acid,
sulfuric acid, nitric acid, and trifluoroacetic acid
(TFA). These acids may each be used singly or may be
221102_G2572WO English Translation (Final) clean copy
used in combination of two or more thereof. A method for
adding the acid may involve adding the acid in the form
of a solution containing the acid dissolved in a solvent
mentioned later.
[0145] The solvent for use in the elimination of the
protective group P' in step 4 of synthesis 1 is not
particularly limited as long as the solvent dissolves the
starting materials to some extent without inhibiting the
reaction. Examples thereof include: water; amides such
as N,N-dimethylformamide, N,N-dimethylacetamide, and N
methylpyrrolidone; halogenated hydrocarbons such as
dichloromethane; aromatic hydrocarbons such as toluene;
ethers such as tetrahydrofuran, diethyl ether,
cyclopentyl methyl ether, 1,4-dioxane, and tert-butyl
methyl ether; alcohols such as methanol, ethanol, n
propanol, and isopropanol; nitriles such as acetonitrile
and propionitrile; and arbitrary mixed solvents thereof.
Water, tetrahydrofuran, dichloromethane, 1,4-dioxane,
methanol, or a mixed solvent prepared by mixing two or
more of them is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually -20°C to 150°C,
preferably 0°C to 100°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 24 hours,
preferably 30 minutes to 10 hours.
221102_G2572WO English Translation (Final) clean copy
[0146] The substituent Rx can be eliminated by adding a
base to a solution containing the compound (1A)-2 or
(2A)-2.
Examples of the base for use in the elimination of
the substituent Rx in step 4 of synthesis 1 include
alkali metal hydroxide such as lithium hydroxide, sodium
hydroxide, and potassium hydroxide, and alkaline earth
metal hydroxide such as calcium hydroxide and magnesium
hydroxide. Alkali metal hydroxide is preferred, and
lithium hydroxide or sodium hydroxide is more preferred.
These bases may each be used singly or may be used in
combination of two or more thereof. A method for adding
the base may involve adding the base in the form of a
solution containing the base dissolved in a solvent
mentioned later.
[0147] The solvent for use in the elimination of the
substituent Rx in step 4 of synthesis 1 is not
particularly limited as long as the solvent dissolves the
starting materials to some extent without inhibiting the
reaction. Examples thereof include: water; amides such
as N,N-dimethylformamide, N,N-dimethylacetamide, and N
methylpyrrolidone; ethers such as tetrahydrofuran,
diethyl ether, cyclopentyl methyl ether, 1,4-dioxane, and
tert-butyl methyl ether; alcohols such as methanol,
ethanol, n-propanol, and isopropanol; nitriles such as
acetonitrile and propionitrile; and arbitrary mixed
solvents thereof. Water, tetrahydrofuran, methanol, or a
221102_G2572WO English Translation (Final) clean copy
mixed solvent prepared by mixing two or more of them is
preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually -20°C to 100°C,
preferably 00C to 50°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 24 hours,
preferably 30 minutes to 10 hours.
[0148] Step 5 of synthesis 1 is the step of reacting the
compound (1A)-3 or (2A)-3 with methyl iodide
(iodomethane) which is a methylating agent in reaction of
the first stage and with diamine of the compound (al) or
an acid-addition salt, etc. thereof in reaction of the
second stage in a solvent to obtain a compound (1A) or
(2A).
The solvent for use at the first stage in step 5 of
synthesis 1 is not particularly limited as long as the
solvent dissolves the starting materials to some extent
without inhibiting the reaction. Examples thereof
include: amides such as N,N-dimethylformamide, N,N
dimethylacetamide, and N-methylpyrrolidone; halogenated
hydrocarbons such as dichloromethane; esters such as
ethyl acetate; hydrocarbons such as dichlorohexane and n
hexane; aromatic hydrocarbons such as toluene; ethers
such as tetrahydrofuran, diethyl ether, cyclopentyl
methyl ether, 1,4-dioxane, and tert-butyl methyl ether;
221102_G2572WO English Translation (Final) clean copy
and arbitrary mixed solvents thereof. N,N
Dimethylformamide or N-methylpyrrolidone is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually 00C to 150°C,
preferably 100C to 1200C, more preferably 200C to 50°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 1 minute to 48 hours,
preferably 10 minutes to 24 hours.
[0149] At the second stage in step 5 of synthesis 1, a
compound (1A) or (2A) is produced through reaction with
diamine of the compound (al) or an acid-addition salt,
etc. thereof in the presence or absence of water. The
compound (al) may be an acid-addition salt, etc.
Examples of the acid-addition salt, etc. include
hydrochloride, sulfate, and acetate.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually 00C to 150°C,
preferably 200C to 120°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 48 hours,
preferably 30 minutes to 24 hours.
[0150] Both the protective group P' and the substituent Rx
in the compound (1A)-2 or (2A)-2 are eliminated in a
solvent in step 4 of synthesis 1, and then, step 5 is
performed to produce a compound (1A) or (2A).
221102_G2572WO English Translation (Final) clean copy
Alternatively, as shown below, the substituent Rx in the
compound (1A)-2 may be eliminated to prepare a compound
(1A)-3' (step 4a), followed by the step 5 mentioned above
to prepare a compound (1A)-4, and then, the protective
group P1 can be eliminated to produce a compound (1A)
(step 4b). The compound (2A) can also be produced by the
same scheme as described below.
[0151]
1) Mel NH2 HN S O HN S O 2) NH 2 HN HNHY HN ORx HN OH (al)
Step 4a R 1 Step 5 N-N N-N, 1
(1A)-2 (1A)-3' Y Y
HN N HN O HN zN HN 0 HN OH HN OH
R Step 4b R N-Np 1 N-NH
(1A)-4 (1A)
[0152] In the scheme, the same methods as in step 4 can
be applied to a method for eliminating the substituent Rx
in step 4a, and a method for eliminating the protective
group P1 in step 4b.
[0153] <Synthesis 2>
In synthesis 1, the compound (1A) or (2A) is
produced using the compound (1A)-1 or (2A)-1 in which the
221102_G2572WO English Translation (Final) clean copy
amino group of the 5-membered heterocyclic ring is
protected with the protective group P1. This compound
may be produced using a compound that is not protected
with the protective group P1.
For example, the compound (1A) may be produced
through each step of synthesis 2 given below using a
compound (1A)-1'. The compound (2A) can also be produced
by the same scheme as in the following synthesis 2.
0 HN S O H 2N ORx CH 3NCS ORx
N-NH Step 3 N-NH Step 4a
(1A)- 1' (1 A)-2' 1) Mel
2)-1 HN S 2 HNH 2 HNN HN 1 OH (al) 2 HN OH
N-NH St 5 R O N-NH (1A)-3 (1A)
[0154] Step 3 and step 5 of synthesis 2 are the same as
step 3 and step 5 of synthesis 1.
Step 4a of synthesis 2 is the step of eliminating
the substituent Rx in the compound (1A)-2' or (2A)-2' in
a solvent to obtain a compound (1A) -3 or (2A) -3. Unlike
step 4 of synthesis 1, only the operation of eliminating
the substituent Rx is performed in step 4a of synthesis
2.
221102_G2572WO English Translation (Final) clean copy
The substituent Rx can be eliminated by adding a
base, as in the method for eliminating the substituent Rx
in step 4 of the synthesis 1 mentioned above. The type
of the base for use in step 4a of synthesis 2 and a
method for adding the base are the same as in the method
for eliminating the substituent Rx in step 4 of the
synthesis 1 mentioned above.
[0155] <Synthesis 3>
The compound (lA)-1 for use as a starting material
in the synthesis 1 mentioned above can be produced
through steps 6 to 8 of synthesis 3 given below. The
compound (2A)-1 can also be produced by the same scheme
as in the following synthesis 3.
0 0 Ph NH
R ~ORX ZR ORX Ph
Step Step N-NH N-N
(1A)-1-1(1A)-1-2 Ph Ph Y 0 0 N ORX H 2N ORX
R1 V3) R1 N--N P1 Step8 N-N
(1A)-1-3 (1A)-1
[0156] In the formulas, Z is a chlorine atom, a bromine
atom, or an iodine atom.
[0157] Step 6 of synthesis 3 is the step of introducing
the protective group P1 to the amino structure moiety of
the 5-membered heterocyclic ring of a compound (1A)-1-1
221102_G2572WO English Translation (Final) clean copy
in a solvent to obtain a compound (1A)-1-2. The compound
(1A)-1-1 is known in the art or may be produced from
another compound known in the art.
A method known in the art can be used as a method
for introducing the protective group P'. The method can
be performed in accordance with, for example, a method
described in Org. Biomol. Chem., 2017, 15, 8614-8626, or
J. Org. Chem., 2015, 80, 7713-7726. Specifically, the
protective group can be introduced to the amino structure
moiety using a compound corresponding to the protective
group to be introduced.
When the protective group PI is, for example, a
tetrahydropyranyl group, the tetrahydropyranyl group can
be introduced to the amino structure moiety of the 5
membered heterocyclic ring through reaction with
dihydropyran (DHP) in the presence of an acid catalyst
(p-toluenesulfonic acid, etc.).
[0158] The solvent for use in step 6 of synthesis 3 is
not particularly limited as long as the solvent dissolves
the starting materials to some extent without inhibiting
the reaction. Examples thereof include: amides such as
N,N-dimethylformamide, N,N-dimethylacetamide, and N
methylpyrrolidone; halogenated hydrocarbons such as
dichloromethane; esters such as ethyl acetate;
hydrocarbons such as dichlorohexane and n-hexane;
aromatic hydrocarbons such as toluene and xylene; ethers
such as tetrahydrofuran, diethyl ether, cyclopentyl
221102_G2572WO English Translation (Final) clean copy
methyl ether, 1,4-dioxane, and tert-butyl methyl ether;
and arbitrary mixed solvents thereof. Dichloromethane,
toluene, xylene, or tetrahydrofuran is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually 0°C to 150°C,
preferably 00C to 50°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 48 hours,
preferably 30 minutes to 24 hours.
[0159] Step 7 of synthesis 3 is the step of reacting the
compound (1A)-1-2 with benzophenone imine in the presence
of a catalyst in a solvent under a stream of an inert gas
to obtain a compound (1A)-1-3.
Examples of the catalyst for use in step 7 of
synthesis 3 include organic palladium complexes such as
tris(dibenzylideneacetone)dipalladium and palladium
acetate.
[0160] In step 7 of synthesis 3, the reaction may be
performed in the presence of a ligand or may be performed
in the absence of a ligand.
Examples of the ligand for use in step 7 of
synthesis 3 include 4,5'-bis(diphenylphosphino)-9,9'
dimethylxanthene (Xantphos) and 2,2'
bis(diphenylphosphino)-1,1'-binaphthyl (BINAP).
221102_G2572WO English Translation (Final) clean copy
[0161] In step 7 of synthesis 3, the reaction may be
performed in the presence of a base or may be performed
in the absence of a base.
Examples of the base for use in step 7 of synthesis
3 include: alkali metal carbonate such as cesium
carbonate, potassium carbonate, sodium carbonate, and
sodium bicarbonate; alkali metal phosphate such as
tribasic potassium phosphate, sodium phosphate, and
sodium biphosphate; alkali metal fluoride such as cesium
fluoride and potassium fluoride; alkylamines such as
triethylamine and N,N-diisopropylethylamine; pyridines
such as pyridine and 4-dimethylaminopyridine; and 1,8
diazabicyclo[5.4.0]-7-undecene. Alkali metal carbonate
is preferred, and cesium carbonate is more preferred.
These bases may each be used singly or may be used in
combination of two or more thereof. A method for adding
the base may involve adding the base in the form of a
solution containing the base dissolved in a solvent
mentioned later.
[0162] The solvent for use in step 7 of synthesis 3 is
not particularly limited as long as the solvent dissolves
the starting materials to some extent without inhibiting
the reaction. Examples thereof include: water; amides
such as N,N-dimethylformamide, N,N-dimethylacetamide, and
N-methylpyrrolidone; halogenated hydrocarbons such as
dichloromethane; esters such as ethyl acetate;
hydrocarbons such as dichlorohexane and n-hexane;
221102_G2572WO English Translation (Final) clean copy
aromatic hydrocarbons such as toluene and xylene; ethers
such as tetrahydrofuran, diethyl ether, cyclopentyl
methyl ether, 1,4-dioxane, and tert-butyl methyl ether;
and arbitrary mixed solvents thereof. Toluene, xylene,
or 1,4-dioxane is preferred.
Examples of the inert gas used include nitrogen gas
and argon gas.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually 500C to 250°C,
preferably 700C to 200°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 1 hour to 48 hours,
preferably 3 hours to 24 hours.
[0163] Step 8 of synthesis 3 is the step of obtaining a
compound (1A)-1 from the compound (1A)-1-3.
A method for obtaining the compound (1A)-1 in step 8
can involve obtaining the compound (1A)-1 through the
reductive reaction of the compound (1A)-1-3 in the
presence of a metal catalyst and in the presence of
hydrogen gas in a solvent.
Examples of the metal catalyst for use in step 8 of
synthesis 3 include inhomogeneous catalysts such as Pd-C
catalysts, palladium hydroxide catalysts, Pt-C catalysts,
and platinum oxide catalysts.
The solvent for use in step 8 of synthesis 3 is not
particularly limited as long as the solvent dissolves the
221102_G2572WO English Translation (Final) clean copy
starting materials to some extent without inhibiting the
reaction. Examples thereof include: water; acetic acid;
amides such as N,N-dimethylformamide, N,N
dimethylacetamide, and N-methylpyrrolidone; esters such
as ethyl acetate; hydrocarbons such as dichlorohexane and
n-hexane; aromatic hydrocarbons such as toluene and
xylene; ethers such as tetrahydrofuran, diethyl ether,
cyclopentyl methyl ether, 1,4-dioxane, and tert-butyl
methyl ether; alcohols such as methanol, ethanol, n
propanol, and isopropanol; nitriles such as acetonitrile
and propionitrile; and arbitrary mixed solvents thereof.
Ethyl acetate, tetrahydrofuran, or methanol is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually 0°C to 150°C,
preferably 200C to 100°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 120
hours, preferably 30 minutes to 48 hours.
[0164] In step 8, the compound (1A)-1 may be obtained by
hydrolyzing the compound (1A)-1-3 by the addition of an
acid (e.g., citric acid) solution to a solution
containing the compound (1A)-1-3.
The solvent for use in step 8 of synthesis 3 is not
particularly limited as long as the solvent dissolves the
starting materials to some extent without inhibiting the
reaction. Examples thereof include: water; amides such
221102_G2572WO English Translation (Final) clean copy
as N,N-dimethylformamide, N,N-dimethylacetamide, and N
methylpyrrolidone; esters such as ethyl acetate;
hydrocarbons such as dichlorohexane and n-hexane;
aromatic hydrocarbons such as toluene and xylene; ethers
such as tetrahydrofuran, diethyl ether, cyclopentyl
methyl ether, 1,4-dioxane, and tert-butyl methyl ether;
alcohols such as methanol, ethanol, n-propanol, and
isopropanol; nitriles such as acetonitrile and
propionitrile; and arbitrary mixed solvents thereof.
Tetrahydrofuran is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually 0°C to 200°C,
preferably 00C to 100°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 120
hours, preferably 30 minutes to 48 hours.
[0165] <Synthesis 4>
The compound (1A)-1 for use as a starting material
in the synthesis 1 mentioned above may be produced
through each step of synthesis 4 given below using a
compound (1A)-1-4 given below, in addition to the
synthesis 3 mentioned above. The compound (2A)-1 can
also be produced by the same scheme as in the following
synthesis 4.
221102_G2572WO English Translation (Final) clean copy
0 0 0 02N ORX 0 2N ORx H2 N ORX
R1 1 R1 3W R1 N-NH Step 9 N-N Step10 N-N. 1
(1 A)- 1-4 (1 A)-1 -5 (1A)- 1
[0166] Step 9 of synthesis 4 is the step of introducing
the protective group P1 to the amino structure moiety of
the 5-membered heterocyclic ring of a compound (1A)-1-4
in a solvent to obtain a compound (1A)-1-5. The compound
(1A)-1-4 is known in the art or may be produced from
another compound known in the art. The same method as in
step 6 of the synthesis 3 mentioned above can be adopted
as a method for introducing the protective group P1 in
step 9. The solvent used is also the same as that used
in step 6 of synthesis 3.
[0167] Step 10 of synthesis 4 is the step of reducing the
nitro group of the compound (1A)-1-5 into an amino group
to obtain a compound (1A)-1.
In step 10 of synthesis 4, a method for reducing the
nitro group of the compound (1A)-1-5 into an amino group
to obtain a compound (1A)-1 can involve reducing the
nitro group of the compound (1A)-1-5 into an amino group
through the reductive reaction of the compound (1A)-1-5
in the presence of a reducing agent such as sodium
dithionite in a solvent to obtain a compound (1A)-1.
The solvent for use in step 10 of synthesis 4 using
a reducing agent is not particularly limited as long as
221102_G2572WO English Translation (Final) clean copy
the solvent dissolves the starting materials to some
extent without inhibiting the reaction. Examples thereof
include: water; acetic acid; amides such as N,N
dimethylformamide, N,N-dimethylacetamide, and N
methylpyrrolidone; esters such as ethyl acetate; ethers
such as tetrahydrofuran, diethyl ether, cyclopentyl
methyl ether, 1,4-dioxane, and tert-butyl methyl ether;
alcohols such as methanol, ethanol, n-propanol, and
isopropanol; nitriles such as acetonitrile and
propionitrile; and arbitrary mixed solvents thereof.
Water or ethanol is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually 0°C to 200°C,
preferably 500C to 150°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 48 hours,
preferably 30 minutes to 24 hours.
[0168] In step 10, the compound (1A)-1 may be obtained by
reducing the nitro group of compound (1A)-1-5 into an
amino group through the reductive reaction of the
compound (1A)-1-5 in the presence of a metal catalyst and
in the presence of hydrogen gas in a solvent.
In this case, examples of the metal catalyst used
include inhomogeneous catalysts such as Pd-C catalysts,
palladium hydroxide catalysts, Pt-C catalysts, and
platinum oxide catalysts.
221102_G2572WO English Translation (Final) clean copy
The solvent for use in step 10 of synthesis 4 using
a metal catalyst is not particularly limited as long as
the solvent dissolves the starting materials to some
extent without inhibiting the reaction. Examples thereof
include: water; acetic acid; amides such as N,N
dimethylformamide, N,N-dimethylacetamide, and N
methylpyrrolidone; esters such as ethyl acetate; ethers
such as tetrahydrofuran, diethyl ether, cyclopentyl
methyl ether, 1,4-dioxane, and tert-butyl methyl ether;
alcohols such as methanol, ethanol, n-propanol, and
isopropanol; nitriles such as acetonitrile and
propionitrile; and arbitrary mixed solvents thereof.
Acetic acid or tetrahydrofuran is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually 0°C to 200°C,
preferably 200C to 100°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 48 hours,
preferably 30 minutes to 24 hours.
[0169] <Synthesis 5>
A compound (4)-S which is an S form of the compound
(4) for use as a starting material in the process 1
mentioned above, wherein R is a C1-C6 alkyl group can be
produced through steps 11 to 13 of synthesis 5 given
below. The compound (4)-S is included in the compound
(4).
221102_G2572WO English Translation (Final) clean copy
0 ||
H2N Step 9 O BrZn CO 2 RStep 10 11 (a4) -S., (a5) A-CHO3 3 Step 11 A Step 12 (4)-1 (4)-S-1
So H2N COR 0 A Step 13 A
(4)-S-2 (4)-S
[0170] Step 11 of synthesis 5 is the step of reacting a
compound (4)-i with (R)-2-methylpropane-2-sulfinamide of
a compound (a4) in the presence of pyrrolidine and a
molecular sieve (4 angstroms) in a solvent to obtain a
compound (4)-S-1.
The solvent for use in step 11 of synthesis 5 is not
particularly limited as long as the solvent dissolves the
starting materials to some extent without inhibiting the
reaction. Examples thereof include: halogenated
hydrocarbons such as dichloromethane (DCM); aromatic
hydrocarbons such as toluene and xylene; ethers such as
tetrahydrofuran, diethyl ether, cyclopentyl methyl ether,
1,4-dioxane, and tert-butyl methyl ether; and arbitrary
mixed solvents thereof. Toluene or tetrahydrofuran is
preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
221102_G2572WO English Translation (Final) clean copy
solvent, and the like, and is usually -20°C to 150°C,
preferably 0°C to 120°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 48 hours,
preferably 30 minutes to 24 hours.
[0171] Step 12 of synthesis 5 is the step of reacting the
compound (4)-S-1 with a compound (a5) in a solvent to
obtain a compound (4)-S-2.
The compound (a5) used is a compound corresponding
to R of the compound (4)-S of interest. The compound
(a5) is known in the art, or can be produced from a
compound known in the art in accordance with the
description of Examples of the present specification
mentioned later.
Specific examples of the compound (a5) include (2
methoxy-2-oxoethyl)zinc(II) bromide, (2-ethoxy-2
oxoethyl)zinc(II) bromide, (2-isopropoxy-2
oxoethyl)zinc(II) bromide, and [2-(tert-butoxy)-2
oxoethyl]zinc(II) bromide.
[0172] The solvent for use in step 12 of synthesis 5 is
not particularly limited as long as the solvent dissolves
the starting materials to some extent without inhibiting
the reaction. Examples thereof include: halogenated
hydrocarbons such as dichloromethane (DCM); aromatic
hydrocarbons such as toluene; ethers such as
tetrahydrofuran, diethyl ether, cyclopentyl methyl ether,
221102_G2572WO English Translation (Final) clean copy
1,4-dioxane, and tert-butyl methyl ether; and arbitrary
mixed solvents thereof. Tetrahydrofuran is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually -78°C to 100°C,
preferably -20°C to 50°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 48 hours,
preferably 30 minutes to 24 hours.
[0173] Step 13 of synthesis 5 is the step of reacting the
compound (4)-S-2 with an acid in a solvent to obtain a
compound (4)-S.
Examples of the acid for use in step 13 of synthesis
include: organic acids such as acetic acid, formic
acid, trifluoroacetic acid, and p-toluenesulfonic acid;
and inorganic acids such as hydrochloric acid,
hydrobromic acid, and sulfuric acid. Hydrochloric acid
is preferred. These acids may each be used singly or may
be used in combination of two or more thereof. A method
for adding the acid may involve adding the acid in the
form of a solution containing the acid dissolved in a
solvent mentioned later.
[0174] The solvent for use in step 13 of synthesis 5 is
not particularly limited as long as the solvent dissolves
the starting materials to some extent without inhibiting
the reaction. Examples thereof include: water; alcohols
such as methanol and ethanol; halogenated hydrocarbons
221102_G2572WO English Translation (Final) clean copy
such as dichloromethane (DCM); aromatic hydrocarbons such
as toluene; ethers such as tetrahydrofuran, diethyl
ether, cyclopentyl methyl ether, 1,4-dioxane, and tert
butyl methyl ether; and arbitrary mixed solvents thereof.
An ether, an alcohol or an arbitrary mixed solvent
thereof is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually -20°C to 100°C,
preferably 00C to 50°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 1 minute to 48 hours,
preferably 10 minutes to 24 hours.
The compound (4)-S thus obtained may be an acid
addition salt, etc. Examples of the acid-addition salt,
etc. include hydrochloride, sulfate, and acetate.
[0175] As described above, the compound (4)-S can be
synthesized through steps 11 to 13 of synthesis 5. An
asymmetric compound opposite to the compound (4)-S can be
synthesized through steps 11 to 13 using, for example,
(S)-2-methylpropane-2-sulfinamide, instead of the
compound (a4). Further, a racemate of the compound (4)-S
can be synthesized through steps 11 to 13 using, for
example, racemic 2-methylpropane-2-sulfinamide, instead
of the compound (a4).
[0176] <Synthesis 6>
221102_G2572WO English Translation (Final) clean copy
The compound (3) for use as a starting material in
the process 1 mentioned above, wherein R is a Ci-C6 alkyl
group can be produced through steps 14 and 15 of the
following synthesis 6.
H 2 p OH H H H 2N CO 2 R (a6) P N CO2R H2N N CO2R A Step 4 0 A Step 15 0 A
(4) (3)-i (3)
[0177] Step 14 of synthesis 6 is the step of reacting the
compound (4) obtained by a method such as synthesis 5 or
an acid-addition salt, etc. thereof with a compound (a6)
in the presence of a dehydration-condensation agent in a
solvent to obtain a compound (3)-1.
In the formulas, P 2 represents a protective group
such as a t-butoxycarbonyl group (Boc).
[0178] Examples of the dehydration-condensation agent for
use in step 14 of synthesis 6 include
dicyclohexylcarbodiimide (DCC), N,N'
diisopropylcarbodiimide (DIC), 1-(3-dimethylaminopropyl)
3-ethylcarbodiimide or hydrochloride thereof (EDC, EDAC),
N,N-dicyclohexylamide, 1-hydroxybenzotriazole,
carbonyldiimidazole, 1H-benzotriazol-1
yloxytris(dimethylamino)phosphonium hexafluorophosphate,
and O-(7-azabenzotriazol-1-yl)-1,1,3,3-tetramethyluronium
hexafluorophosphate. 1-(3-Dimethylaminopropyl)-3
ethylcarbodiimide or hydrochloride thereof (EDC, EDAC) is
preferred.
221102_G2572WO English Translation (Final) clean copy
[0179] In step 14 of synthesis 6, the reaction may be
performed in the presence of a base or may be performed
in the absence of a base.
Examples of the base for use in step 14 of synthesis
6 include N,N-diisopropylethylamine, N-methylmorpholine,
triethylamine, 4-(N,N-dimethylamino)pyridine, and N
ethyl-N-isopropylpropan-2-amine. N,N
Diisopropylethylamine or triethylamine is preferred.
These bases may each be used singly or may be used in
combination of two or more thereof. A method for adding
the base may involve adding the base in the form of a
solution containing the base dissolved in a solvent
mentioned later.
[0180] The solvent for use in step 14 of synthesis 6 is
not particularly limited as long as the solvent dissolves
the starting materials to some extent without inhibiting
the reaction. Examples thereof include: amides such as
N,N-dimethylformamide (DMF), N,N-dimethylacetamide, and
N-methylpyrrolidone; halogenated hydrocarbons such as
dichloromethane (DCM); esters such as ethyl acetate;
hydrocarbons such as dichlorohexane and n-hexane;
aromatic hydrocarbons such as toluene; ethers such as
tetrahydrofuran, diethyl ether, cyclopentyl methyl ether,
1,4-dioxane, and tert-butyl methyl ether; and arbitrary
mixed solvents thereof. Dichloromethane is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
221102_G2572WO English Translation (Final) clean copy
solvent, and the like, and is usually -20°C to 100°C,
preferably 00C to 50°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 72 hours,
preferably 30 minutes to 48 hours.
[0181] Step 15 of synthesis 6 is the step of deprotecting
the protective group P 2 from the compound (3)-1 using an
acid in a solvent to obtain a compound (3).
Examples of the acid for use in step 15 of synthesis
6 include: organic acids such as acetic acid, formic
acid, trifluoroacetic acid, and p-toluenesulfonic acid;
and inorganic acids such as hydrochloric acid,
hydrobromic acid, and sulfuric acid. Hydrochloric acid
is preferred. These acids may each be used singly or may
be used in combination of two or more thereof. A method
for adding the acid may involve adding the acid in the
form of a solution containing the acid dissolved in a
solvent mentioned later.
[0182] The solvent for use in step 15 of synthesis 6 is
not particularly limited as long as the solvent dissolves
the starting materials to some extent without inhibiting
the reaction. Examples thereof include: water; alcohols
such as methanol and ethanol; halogenated hydrocarbons
such as dichloromethane (DCM); aromatic hydrocarbons such
as benzene, toluene, and xylene; ethers such as
tetrahydrofuran, diethyl ether, cyclopentyl methyl ether,
1,4-dioxane, and tert-butyl methyl ether; nitriles such
221102_G2572WO English Translation (Final) clean copy
as acetonitrile and propionitrile; and arbitrary mixed
solvents thereof. An ether, an alcohol or an arbitrary
mixed solvent thereof is preferred.
The reaction temperature differs depending on the
types, usages, etc. of the starting materials, the
solvent, and the like, and is usually -20°C to 100°C,
preferably 00C to 50°C.
The reaction time differs depending on the reaction
temperature, etc., and is usually 10 minutes to 72 hours,
preferably 30 minutes to 48 hours.
The compound (3) thus obtained may be an acid
addition salt, etc. Examples of the acid-addition salt,
etc. include hydrochloride, sulfate, and acetate.
[0183] In the present specification, the obtained
compound in each step may be isolated or purified by an
approach known in the art or may be subjected directly to
a subsequent step. The isolation or the purification can
be carried out by use of a usual operation, for example,
filtration, extraction, crystallization, or each column
chromatography.
[0184] <Intermediate of present invention>
In one embodiment of the present invention,
compounds given below which are intermediates of the
compound (I) or (II) or a pharmaceutically acceptable
salt thereof are also included in the present invention.
In one embodiment, examples of the intermediate of
the present invention include compounds represented by
221102_G2572WO English Translation (Final) clean copy
the following general formula (X-1) or (X-2) and salts
thereof:
RY 0 RY 0 HN HN HN ORXA HN ORXA
N-N Rz Rz' N-N
(X-1) (X-2)
In the formulas (X-1) and (X-2), RY is a group
represented by the following formula (x)-i or (x)-ii:
Y
HN 1-N HN S
(x)-i (x)-ii wherein Y is a hydrogen atom, a fluorine atom or a
hydroxy group, as in the general formulas (I) and (II),
R7 is a C1-C6 alkyl group, and * represents a binding
position.
R' is a hydrogen atom or a halogen atom, as in the
general formulas (I) and (II).
RXA is a hydrogen atom or a hydrocarbon group having
1 to 6 carbon atoms and is preferably a hydrogen atom or
a C1-C6 alkyl group.
221102_G2572WO English Translation (Final) clean copy
Rz is a hydrogen atom or P' (wherein P' represents,
as mentioned above, a protective group such as a
tetrahydropyranyl (THP) group).
[0185] In one embodiment, RY in the general formula (X-1)
is a group represented by the formula (x)-i.
In one embodiment, RY in the general formula (X-1)
is a group represented by the formula (x)-ii.
In one embodiment, RY in the general formula (X-2)
is a group represented by the formula (x)-i.
In one embodiment, RY in the general formula (X-2)
is a group represented by the formula (x)-ii.
[0186] In one embodiment, examples of the intermediate of
the present invention include compounds represented by
the following general formula (X-1-1) or (X-2-1) and
salts thereof:
RYA O RYA O HN ORXA1 HN ORXA1
R1A R 1A
N-N N-N R RZA
(X-1-1) (X-2-1) In the formulas (X-1-1) and (X-2-1), RYA is a group
represented by the following formula (x)-i-1, (x)-i-2,
(x)-i-3, or (x)-ii-1:
221102_G2572WO English Translation (Final) clean copy
F OH HN N HN N HN N HN S * * *
* (x)-i-1 (x)-i-2 (x)-i-3 ()i wherein * represents a binding position.
RlA is a hydrogen atom, a fluorine atom, or a
chlorine atom.
RXAl is a hydrogen atom or a methyl group.
RZA is a hydrogen atom or a tetrahydropyranyl (THP)
group.
[0187] In one embodiment, RYA in the general formula (X-1
1) is a group represented by the formula (x)-i-1, the
formula (x)-i-2, or the formula (x)-i-3.
In one embodiment, RYA in the general formula (X-1-1)
is a group represented by the formula (x)-ii-1.
In one embodiment, RYA in the general formula (X-2-1)
is a group represented by the formula (x)-i-1, the
formula (x)-i-2, or the formula (x)-i-3.
In one embodiment, RYA in the general formula (X-2-1)
is a group represented by the formula (x)-ii-1.
[0188] In one embodiment, examples of the intermediate of
the present invention include compounds selected from the
following group and salts thereof:
221102_G2572W0 English Translation (Final) clean copy
HN 'rS HN
HN ~ N HN S
FN N~-H HNSHH
HN SHN, HN.,S
N-N N-NN HN X C1 F N-HN-NH N-NH
HN N HNNSNNH HN 0 T 0 HN-SNN H OHHN CHHN OH 0 T N~ H O N O "OH HNH HN~OH C1F FP N-NH N-NO N-NHn N-HCNHNN
00 F
HN S0N~ HN N
01HN11I OH [I OH
[0189] In one embodiment, the intermediate of the present
invention is a compound selected from the following group
or a salt thereof:
HN 1 S .- rN S H 0, HN S HN HN HN aa0 I FoHNl HN 11 HN 01
N-N N-NO N-N C1 F N-NH N-NH N-NH
HN HNS HN S 00 HN S HN SHN S HN OH OH OH HN OH HN 0 OHH0
K11 F KIl KIl OH HO HN OH
0 N-NO N-Nn N-NO C1 F
0 NH N-NH N-NH
221102_G2572WO English Translation (Final) clean copy
[0190] In one embodiment, the intermediate of the present
invention is a compound selected from the following group
or a salt thereof:
F F F F OH
HNrN O HN N O HNrN O HN N O HNrN O HNrN O
OH H OH HN OH HN OH HN OH ci OH F C,: F \ A F P N-NH N-N N-NH N-NH N-NH N-NH
[0191] In one embodiment, the intermediate of the present
invention is a compound selected from the following group
or a salt thereof:
F HN S O HN S HN N HN O HN OH HN OH HN-N HN-N HN-N
[0192] In another embodiment, examples of the
intermediate of the present invention include compounds
represented by the following general formula (X-3) and
salts thereof:
H RW N NC2 H CO 2 R 0 A (X-3)
In the formula (X-3), A is a C6-C1O aryl group or a
heteroaryl group, wherein at least one hydrogen atom of
the aryl group or the heteroaryl group is optionally
221102_G2572WO English Translation (Final) clean copy
replaced with a substituent selected from the group
consisting of a halogen atom, a hydroxy group, a C1-C6
alkyl group, a C1-C6 haloalkyl group, a C2-C6 alkenyl
group, a C2-C6 alkynyl group, a C1-C6 alkoxy group, a C1-C6
haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
cycloalkenyl group, a C3-C6 cycloalkoxy group, a
heterocyclyl group, a heteroaryl group optionally
substituted by a C1-C6 alkyl group, a cyano group, a
carboxyl group, a carbamoyl group, a C1-C6 alkoxycarbonyl
group, a C1-C6 alkylsulfanyl group, and a C1-C6
alkylsulfonyl group, as in the general formulas (I) and
(II).
R is a hydrogen atom or a C1-C6 alkyl group, as in
the general formulas (I) and (II).
Rw is a hydrogen atom or P 2 , wherein P 2 represents,
as mentioned above, a protective group such as a t
butoxycarbonyl group (Boc).
[0193] In one embodiment, examples of the intermediate of
the present invention include compounds represented by
the following general formula (X-3-1) and salts thereof:
H N RW H CO 2 R 0 R2 (X-3-1)
R5 R3
R4
In the formula (X-3-1),
221102_G2572WO English Translation (Final) clean copy
R 2 is a hydrogen atom or a halogen atom,
R3 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
C1-C6 haloalkoxy group, a C3-Cs6 cycloalkyl group, a C3-Cs6
cycloalkoxy group, a heterocyclyl group, or a heteroaryl
group optionally substituted by a C1-C6 alkyl group,
R4 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, or a
C1-C6 haloalkoxy group,
R5 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
C1-C6 haloalkoxy group, or a heteroaryl group optionally
substituted by a C1-C6 alkyl group,
R 6 is a hydrogen atom or a halogen atom,
R is a hydrogen atom or a C1-C6 alkyl group, as in
the general formulas (I) and (II), and
Rw is a hydrogen atom or P 2 (wherein P 2 represents,
as mentioned above, a protective group such as a t
butoxycarbonyl group (Boc)).
[0194] In one embodiment, examples of the intermediate of
the present invention include compounds represented by
the following general formula (X-3-2) and salts thereof:
H RW -1N Rw N NCO 2 R 0 A1 (X-3-2)
221102_G2572WO English Translation (Final) clean copy
R is a hydrogen atom or a C1-C6 alkyl group, as in
the general formulas (I) and (II).
Rw is a hydrogen atom or P 2 (wherein P 2 represents,
as mentioned above, a protective group such as a t
butoxycarbonyl group (Boc)).
A' is a group selected from the group given below.
In the following formulas, * represents a binding
position.
C1 CF 3 Br CF 3 I CF3 CF 3 O CF 3 F CF3 F 3C CF 3
F CI F C
F3C N CF 3 CF 3 CF 3 CF 3 CF 3 CF3 NQ~> F
Cl CF3 Br Cl Cl Br Cl Br Br I Br Cl i F OMe
F __:tF F_[b C1 F Br F 2HCO CF 3 CI OCHF 2 Br OCHF 2 OCHF 2 OCHF 2 F
F 2HCO N F2 HCO OCHF 2 Cl OCF 3 Br OCF3 OCF 3 OCF 3 CF 3 F
OCF 3 OCHF 2 CHF 2 O Cl Br
N OCHF 2 CF3
[0195] In one embodiment, the intermediate of the present
invention may be a salt of the compound represented by
any of the formulas (X-1) to (X-3), the formula (X-1-1),
the formula (X-2-1), the formula (X-3-1) and the formula
221102_G2572WO English Translation (Final) clean copy
(X-2-3). Examples of the salt include, but are not
particularly limited to, the "acid-addition salt, etc."
and the "base-addition salt, etc." listed as examples of
the "pharmaceutically acceptable salt" mentioned above.
[0196] All references including publications, patent
applications and patents cited herein are incorporated
herein by reference in their entirety.
Examples
[0197] Hereinafter, the present invention will be
described in more detail with reference to Reference
Examples, Examples and Test Examples. However, the scope
of the present invention is not limited by these
examples. The names Reference Examples, Examples and
Test Examples are merely given for the sake of
convenience in order to discriminate among experimental
examples and do not mean that any of these examples are
not included in the present invention.
[0198] DUIS in the ionization mode of a mass spectrum is
a mixed mode of ESI and APCI.
[0199] In the present specification, 'H-NMR is indicated
by chemical shift (6) with tetramethylsilane as an
internal standard (0 ppm), and a coupling constant (J
value) is indicated by Hz unit, unless otherwise
specified. The abbreviation of the splitting pattern of
each peak means the following: s: singlet, d: doublet,
dd: double doublet, ddd: double double doublet, t:
221102_G2572WO English Translation (Final) clean copy
triplet, tt: triple triplet, quin: quintet, and m:
multiplet.
[0200] Abbreviations described in Reference Examples,
Examples, Test Examples and chemical structural formulas
are typically used in meanings that are generally used in
the field of organic chemistry or pharmaceuticals, and
specifically, should be understood as the abbreviations
of the following terms by those skilled in the art.
Me: methyl group
DMSO: dimethyl sulfoxide
tert: tertiary
N: normal
M: molar concentration
ESI: electrospray ionization
APCI: atmospheric pressure chemical ionization
EI: electronic ionization
CI: chemical ionization
[0201] Reference Example 1-(a)
Production of (R,E)-N-(3-chloro-5
(trifluoromethyl)benzylidene)-2-methylpropane-2
sulfinamide
N
CI F3
[0202] To a solution of 3.82 g of 3-chloro-5
(trifluoromethyl)benzaldehyde in toluene (20 ml), 2.35 g
221102_G2572WO English Translation (Final) clean copy
of (R)-2-methylpropane-2-sulfinamide (manufactured by
Combi-Blocks Inc.), 0.079 ml of pyrrolidine, and 4 g of a
molecular sieve (4 angstroms) were added at room
temperature under a stream of argon, and the mixture was
stirred at 700C for 2.5 hours.
After the completion of reaction, the reaction mixture
was filtered through celite and concentrated under
reduced pressure. Hexane was added to the concentration
residue, and the mixture was stirred at room temperature
for 30 minutes. A deposited solid was collected by
filtration, washed with hexane, and then dried under
reduced pressure at 600C to obtain 2.22 g of the title
compound as a white solid.
Mass spectrum (ESI, m/z): 312 [M+H]+.
[0203] Reference Example 2-(a), etc.:
Corresponding starting compounds were treated in the
same manner as in Reference Example 1-(a) to obtain
compounds described in the following Tables 1-1 to 1-8.
221102_G2572WO English Translation (Final) clean copy
[0204]
Table 1-1 Reference Structural formula Mass spectrum
example2-(a) (Cl, m/z): 356, 358 [M+H]+
Br, CF 3
Exaplen3-(a) (ESI, m/z): 404 [M+H]+
Example 4-(a) (DUIS,m/z):292[M+H]+
S CF 3
eape5-rc (ESI, m/z): 308 [M+H]+
CF 3
s O F, CF 3 Exaple)eFC (ESI, m/z): 296 [M+H]+
eame°ea)F (ESI, m/z): 346 [M+H]+
F-1 3 CF
221102_G2572WO English Translation (Final) clean copy
[ 02 0 5]
Table 1-2 Reference Example Structural formula Mass spectrum
Reference Example N (ESI, m/z): 296 [M+H]+ 9-(a) F
CF 3
Reference Example N c (ESI, m/z): 312 [M+H]+ 1O0-(a)I
CF 3
Reference Example N (ESI, m/z): 296 [M+H]+ 11-(a) F
F 3C
Reference Example F N (ESI, m/z): 312 [M+H]+ 12-(a) Exapl C
Reference Example ESI, m/z): 292 [M+H]+ 13-(a)
C;F, F
ReferenceExample N (ESI,mlz):292 [M+H] 14-(a) CF 3
221102_G2572WO English Translation (Final) clean copy
[0206]
Table 1-3 Reference Example Structural formula Mass spectrum
>S-1
Reference Example NESI, m/z): 330 [M+H]+. 15-(c) (EImz:30MH~ CI CF 3 F
Reference Example N (ESI, m/z): 318, 320 [M+H]+ 16-(a) Br OMe
Reference Example N DUIS, m/z): 278 [M+H]+ 17-(a)
CI CI
Reference Example N (ESI, m/z): 322, 324 [M+H]+ 18-(a)
Br CI
Reference Example NESI, m/z): 368 [M+H]+ 19-(a) r& Br BBr
>L..10 Reference Example N (ESI, m/z): 414, 416 [M+H]+ 20-(a)
1 Br
221102_G2572WO English Translation (Final) clean copy
[0207]
Table 1-4 Reference Example Structural formula Mass spectrum
Reference Example NESI, m/z): 370 [M+H]+ 21 -(c)
CI I
Reference Example N (ESI, m/z): 262 [M+H]+ 22-(a) CI F
Reference Example F (ESI, m/z): 306, 308 [M+H]+
23-(a) Br& F
Reference Example N ESI, m/z): 344 [M+H]+ F
F 0& F3 O 25(b C N" (ESI, mlz): 344[M+H]+ FF
Reference Example NE
F 0 OC F
Reference Example Brl 0 F N (ESI, m/z): 354, 356 [M+H]+ 26-(b) F
Br 0 F
221102_G2572WO English Translation (Final) clean copy
[ 02 0 8]
Table 1-5 Reference Example Structural formula Mass spectrum
Reference Example (ESI, m/z): 294 [M+H]+ 27-(b) F
O F
Reference Example N (ESI, m/z): 294 [M+H]+ 28-(b)F F
F 0
Reference Example (Cl, m/z): 342 [M+H+ 30-(c) F F F 0 0 F
Reference Example N (ESI, m/z): 328 [M+H]+ 31-(a)
CI OCF3
Reference Example NESI, m/z): 372, 374 [M+H+ 32-(a)
Br OCF 3
Reference Example NESI, m/z): 312 [M+H]+ 33-(a) F
F 3CO
221102_G2572WO English Translation (Final) clean copy
[ 02 0 9 ]
Table 1-6 Reference Example Structural formula Mass spectrum
Reference Example NESI, m/z): 312 [M+H]+. 34-(a) OCF 3 F
Reference Example N (ESI, m/z): 278 [M+H]+ 35-(a)
CF 3
Reference Example N (ESI, m/z): 294 [M+H]+ 36-(a) 6 OF OCF 3
Reference Example (ESI, m/z): 276 [M+H]+ 37-(a) F
0 F
Reference Example N (ESI, m/z): 260 [M+H]+ - F F
Reference Example N (ESI, mlz): 250 [M+H]+ 39-(a)N
221102_G2572WO English Translation (Final) clean copy
[0210]
Table 1-7 Reference Example Structural formula Mass spectrum
s~ Reference Example N (ESI, m/z): 266 [M+H]+ 40-(a)
0A
Reference Example NESI, m/z): 244 [M+H]+ 41 -(a)
CCI
Reference Example (ESI, m/z): 288, 290 [M+H]+ 42-(a)
BBr
Reference Example N (ESI, m/z): 276 [M+H]+ 43-(a) Ex eESI,m/z):2 N
[M+H]+
N Reference Example NEI /) 34[+] 44-(a) IEI lz:04M+]
s
Reference Example ESI, m/z): 295 [M+H]+ 45-(a) (E&
221102_G2572WO English Translation (Final) clean copy
[ 0 211]
Table 1-8 Reference Example Structural formula Mass spectrum
N Reference Example (ESI, m/z): 276 [M+H]+ 46-(a) O< F
F
Ns Reference Example (ESI, m/z): 278 [M+H]+ 47-(a)I
CF 3
[0212] Reference Example 1-(b)
Production of methyl (S)-3-(((R)-tert
butylsulfinyl)amino)-3-(3-chloro-5
(trifluoromethyl)phenyl)propanoate
1 0
C1 CFj
[0213] To 33.3 g of zinc, 5.05 g of copper(I) chloride
was added with stirring under a stream of argon, followed
by deaeration under reduced pressure at a bath
temperature of 70°C for 15 minutes. After purging with
argon, 45 ml of tetrahydrofuran was added thereto under a
stream of argon, and the mixture was stirred at 700C for
1 hour. Then, the bath temperature was lowered to 40°C,
and 10.4 ml of methyl bromoacetate was added dropwise
221102_G2572WO English Translation (Final) clean copy
thereto at an internal temperature of 300C to 370C. After
the completion of addition, the mixture was stirred at a
bath temperature of 50°C for 1 hour. The reaction
mixture was brought back to room temperature and then
cooled in an ice salt bath. A solution of 5.00 g of
(R,E)-N-(3-chloro-5-(trifluoromethyl)benzylidene)-2
methylpropane-2-sulfinamide synthesized in the same
manner as in Reference Example 1-(a) in tetrahydrofuran
(22.5 ml) was added dropwise thereto at an internal
temperature of -5°C or lower under a stream of argon, and
the mixture was further washed repeatedly with 7.50 ml of
tetrahydrofuran. After the completion of dropwise
addition, the mixture was cooled in an ice bath and
stirred for 2 hours.
After the completion of reaction, the reaction
mixture was filtered through celite (545) and washed with
ethyl acetate. Then, a saturated aqueous solution of
ammonium chloride was added to the filtrate, and the
mixture was stirred. This mixed solution was filtered,
and the filtrate was separated into organic and aqueous
layers. The organic layer was washed with saturated
saline, dried over anhydrous magnesium sulfate, filtered,
and concentrated under reduced pressure. The obtained
concentration residue was purified by medium-pressure
preparative chromatography (silica gel, eluting solvent:
hexane:ethyl acetate) to obtain 4.82 g of the title
compound as a pale yellow oil.
221102_G2572WO English Translation (Final) clean copy
Mass spectrum (ESI, m/z): 386 [M+H]+.
[0214] Reference Example 2-(b), etc.:
Corresponding starting compounds were treated in the
same manner as in Reference Example 1-(b) to obtain
compounds described in the following Tables 2-1 to 2-8.
221102_G2572WO English Translation (Final) clean copy
[ 0 215]
Table 2-1 Reference Structural formula Mass spectrum
Reference HN O (Cl, m/z): 430, 432 [M+H]+ Example 2-(b)0 1 Br CF 3
>L%.0 Reference HN O (ESI, m/z): 478 [M+H]+ Example 3-(b)0 1 1 CF 3
> s1 Reference HN 4b (ESI, m/z): 366 [M+H]+ Example 4-(b)0
CF 3
> s1 Reference HN (ESI, m/z): 382 [M+H]+ Example 5-(b)0 1 O CF 3
Reference HN O (ESI, m/z): 370 [M+H]+ Example 6-(b) ½ F CF 3
R efnce HN (ESI, m/z): 420 [M+H]+ Example 7-(b)½
F 3C CF 3
221102_G2572WO English Translation (Final) clean copy
[ 0 216]
Table 2-2 Reference Example Structural formula Mass spectrum
Reference Example HN CO 2 Me (ESI, m/z): 370 [M+H]+ 9-(b) F
CF 3
Reference Example HN CC 2 Me (ESI, m/z): 386 [M+H]+ 1 0-(b)N
CF 3
Reference Example HN CO 2 Me (ESI, m/z): 370 [M+H]+
F 3C
Reference Example HN CO2MO (ESI, m/z): 386 [M+H]+ 12-(b) CM
F 3C
Reference Example H(ESI, m/z): 370 [M+H]+ 13-(b)CF CF3 F
Reference Example HN O (ESI, m/z): 366 [M+H]+ 14-(b) (
AFCF 3
221102_G2572WO English Translation (Final) clean copy
[0217]
Table 2-3 Reference Example Structural formula Mass spectrum
>KO Reference Example HN O (ESI, m/z): 404 [M+H]+ 15-(d) 0 CI CF 3 F
Reference Example HN CO 2 Me (ESI, m/z): 392, 394 [M+H]+ 16-(b) Br: OMe
Reference Example HN 0 (ESI, m/z): 352 [M+H]+ 17-(b)0
CI C
Reference Example HN O (ESI, m/z): 396, 398 [M+H]+ 18-(b)
Br CI
Reference Example HO (ESI,mz):442[M+H]+ 19-(b)
Br Br
Reference Example HN OMe (ESI,m/z):488,490[M+H] 20-(b)0
I Br
221102_G2572WO English Translation (Final) clean copy
[ 0 218 ]
Table 2-4 Reference Example Structural formula Mass spectrum
Reference Example HN (ESI, m/z): 444 [M+H]+ 21 -(d)0
CI I
Reference Example HN CQ 2Me (ESI, m/z): 336 [M+H]+ 22-(b)F
Cl
Reference Example H CO2Me (ESI, m/z): 380, 382 [M+H]+ 23-(b) COMFEImz:8,8[+]
Br
Reference Example HN 0¾ (ESI, m/z): 418 [M+H]+ 24-(c) F F 0 CF 3
Reference Example HN o ESI, m/z): 384 [M+H]+ 25-(c) H F
C1 O1j F
Reference Example HN (ESI, m/z): 428,430 [M+H]+ 26-(c) 0 F 1 Br O F
221102_G2572WO English Translation (Final) clean copy
[ 0 219 ]
Table 2-5 Reference Example Structural formula Mass spectrum
Reference Example HN (ESI, m/z): 368 [M+H]+ 27-(c) I . F O F
> S',
Reference Example HN O (ESI, m/z): 46 [M+H]+ 28-(c) FOM EImz:6[+] F F
Reference Example HN (ESI, m/z): 416 [M+H]+ 30-(d) F
F 0 0 F CI CF
Reference Example HN CO2M (ESI, m/z): 402 [M+H]+ 31-(b) 0
CO 00F 3
sz Reference-Example HN (ESI,mz):446,448[M+H] 32-(b) 0
Br 00F 3
>0s Reference Example HN CO 2Me (ESI, m/z): 386 [M+H]+ 33-(b) F
221102_G2572WO English Translation (Final) clean copy
[0220]
Table 2-6 Reference Example Structural formula Mass spectrum
Reference Example HN O (ESI, m/z): 386 [M+H]+ 34-(b) E
OCF 3 F
Reference Example HN (ESI, m/z): 352 [M+H]+ 35-(b) 0
CF 3
Reference Example HN CO 2Ms (ESI, m/z): 368 [M+H]+ 36-(b)
OCF 3
Reference Example HN CO 2Me (ESI, m/z): 350 [M+H]+ 37-(b) F
0F
Reference Example H O (ESI, m/z): 334 [M+H]+ 38-(b) F F
Reference Example HN 01 (ESI, m/z): 324 [M+H]+ 39-(b) (
221102_G2572WO English Translation (Final) clean copy
[ 0 2 21]
Table 2-7 Reference Example Structural formula Mass spectrum
Reference Example HN O (ESI, m/z): 340 [M+H]+ 40-(b) O
Reference Example HN (E0mz:1[+]
Cl
Reference Example HN (ESI,m/z):362,364[M+H]+ 42-(b)
Br
Reference Example HN (ESI, m/z): 350 [M+H]+ 43-(b) 0 N
Reference Example HN (ESI, m/z): 378 [M+H]+ 44-(b) MIENml)38[+]
Reference Example HN (ESI, m/z): 369 [M+H]+ 45-(b) 0(EImz36[+] N 0
221102_G2572WO English Translation (Final) clean copy
[0222]
Table 2-8 Reference Example Structural formula Mass spectrum
HN_,
, Reference Example O (ESI, m/z): 350 [M+H]+ 46-(b) (EImz35[+]
F
Reference Example HN o (ESI, m/z): 352 [M+H]+ 47-(b) 0(EImz35[+]
CF 3
[0223] Reference Example 1-(c)
Production of methyl (S)-3-amino-3-(3-chloro-5
(trifluoromethyl)phenyl)propanoate hydrochloride
HCI H 2N Os
Ci CFa
[0224] To a solution of 24.6 g of methyl (S)-3-(((R)
tert-butylsulfinyl)amino)-3-(3-chloro-5
(trifluoromethyl)phenyl)propanoate synthesized in the
same manner as in Reference Example 1-(b) in cyclopentyl
methyl ether (270 ml), 2.52 ml of methanol and 34.2 ml of
a 4 M solution of hydrogen chloride in cyclopentyl methyl
ether were added at room temperature under a stream of
argon, and the mixture was stirred at room temperature
for 1 hour.
221102_G2572WO English Translation (Final) clean copy
After the completion of reaction, the reaction
mixture was concentrated under reduced pressure. 50 ml
of cyclopentyl methyl ether and 600 ml of hexane were
added to the concentration residue, and the mixture was
stirred at room temperature for 1 hour. A deposited
solid was collected by filtration, washed with hexane,
and then dried under reduced pressure at 500C to obtain
17.5 g of the title compound as a white solid.
Mass spectrum (ESI, m/z): 282 [M+H]+.
[0225] Reference Example 2-(c), etc.:
Corresponding starting compounds were treated in the
same manner as in Reference Example 1-(c) to obtain
compounds described in the following Tables 3-1 to 3-7.
221102_G2572WO English Translation (Final) clean copy
[0226]
Table 3-1 Reference Structural formula Mass spectrum Example HCI
Exampe2-(c) H (Cl, m/z): 326, 328 [M+H]+
Br CF 3
HCI
xarpe3-(c) H2N O (ESI, m/z): 374 [M+H]+
1 CF 3 HCI
Exarple 4-(c) H O1 (DUIS, m/z): 262 [M+H]+
CF 3
HCI
Reference H 2N 02 Example 5-(c) (ESI,mlz):278[M+H]+
O CF3
HCI
exaple6-(c) H2N O (ESI, m/z): 266 [M+H]+
F CF 3
HCI
Reference H 2N CO 2 Me (ESI, m/z): 266 [M+H]+ Example 9-(c) IF(EImz26[H] CF 3
221102_G2572WO English Translation (Final) clean copy
[0227]
Table 3-2 Reference Example Structural formula Mass spectrum HCI
Reference Example H2N CO 2 Me (ESI, m/z): 282 [M+H]+ 10-(c) Cl I1~i CF 3
HCI
Reference Example H 2N CO 2Me (ESI, m/z): 266 [M+H]+ 11-(c) F
F 3C
HCI
Reference Example H 2N CO 2Me (ESI, m/z): 282 [M+H]+ 12-(c) Cl(EImz28M+]
F 3C HCI H 2N Reference Example O 13-(c) 13-c)(ESI, mlz): 266 [M+H+ CF 3 F
HCI H 2N Os Reference Example 0 ESI, m/z): 300 [M+H]+ 15-(e) E CI CF 3 F
HCI H 2N CO 2Me Reference Example ESI, m/z): 288,290 [M+H]+ 16-(c) Br OMe
221102_G2572WO English Translation (Final) clean copy
[ 02 2 8]
Table 3-3 Reference Example Structural formula Mass spectrum
HCI
Reference Example H2N 01 (DUIS, m/z): 248 [M+H]+ 17-(c)
( Cl Cl
HCI
Reference Example H 2N 0 (ESI, m/z): 338 [M+H]+ 19-(c) ( Br Br
HCI
Reference Example H2N OMe (ESI, m/z): 384, 386 [M+H]+ 20-(c) (E0mz:8,8[+]
1Br
HCI
Reference Example H 2N Os (ESI, m/z): 340 [M+H]+ 21-(e) (
CI I HCI
Reference Example H2N CO 2Me (ESI, m/z): 232 [M+H]+ 22-(c) N.F
Cl
Reference Example H 2N CO 2 Me 23-(c) I. F (ESI, mlz): 276,278 [M+H+
BrT
221102_G2572WO English Translation (Final) clean copy
[ 02 2 9 ]
Table 3-4 Reference Example Structural formula Mass spectrum
HCI
Reference Example H2N 0 (ESI, m/z): 314 [M+H]+ 24-(d) F
( F O CF 3
HCI
Reference Example H2N (ESI, m/z): 280 [M+H+ 25-(d) F (ESI,mlz): 2 [M+H]
Cl 0 F
HCI
Reference Example H2N ESI, m/z): 26 [M+H+ 26-(d) F ( Br 0 F HCI H 2N O F 011 Reference Example H (ESI, m/z): 32 [M+H]+ 27-(d) F F F F
Reference Example HN CO 2Me (ESI, mlz): 264 [M+H]+ 28-(d) F< F
HOP F O, O0 Reference Example H2N 0w (ESI, mlz): 312 [M+H+ 30-(e) F N
FA 0 F
221102_G2572WO English Translation (Final) clean copy
[0230]
Table 3-5 Reference Example Structural formula Mass spectrum HCI H 2N O Reference Example O (ESI, m/z): 298 [M+H]+ 31 -(c) 0EImz:9[+]
CI OCF 3
HCI
Reference Example H 2N (ESI, m/z): 342,344 [M+H]+ 32-(c) (E0mz:4,4[+]
Br OCF 3
Reference Example H 2N CO2 Me ESI, m/z): 282 [M+H]+ 33-(c) F(EImz28[+]
F 3CO
HCI
Reference Example H 2N Os (ESI, m/z): 248 [M+H]+ 35-(c) 0(EImz24[+]
CF 3 HCI
Reference Example H 2N CO 2Me (ESI, m/z): 264 [M+H]+ 36-(c) OCF 3
H CI
Reference Example C0 2 Me (ESI, m/z): 246 [M+H+ 37-(c) F 0 F
221102_G2572WO English Translation (Final) clean copy
[ 0 2 31]
Table 3-6 Reference Example Structural formula Mass spectrum
HCI
Reference Example H2N 0 (ESI, m/z): 220 [M+H]+ 38-(c) E (ESI,m/z):230[M+H]+
0 F F
HCI
Reference Example H 2N E m 2[
39-(c) 0 (ESI,mlz):220[M+H]+
HCI
Reference Example H2 NESI, m/z): 2 [M+H+ 40-(c) E
HCI H2N O Reference Example o ESI, m/z): 26 [M+H]+ 43-(c) (
HCI H 2N 0 Reference Example 0 (ESI, mlz): 274 [M+H]+ 44-(c) N
HOI H 2N 0 Reference Example 0(Smz:25[+l 45-(c)(EImz26[+] N O
221102_G2572WO English Translation (Final) clean copy
[0232]
Table 3-7 Reference Example Structural formula Mass spectrum HCI
H2 N O
Reference Example o (ESI, m/z): 246 [M+H]+ 46-(c) 0 F F
HCI
Reference Example 0 ESI, m/z): 248 [M+H]+ 47-(c)(EImz24[+]
CF 3
[0233] Reference Example 1-(d)
Production of methyl (S)-3-(2-((tert
butoxycarbonyl)amino)acetamido)-3-(3-chloro-5
(trifluoromethyl)phenyl)propanoate
0 0
Cl 'CF2
[0234] To a solution of 2.23 g of methyl (S)-3-amino-3
(3-chloro-5-(trifluoromethyl)phenyl)propanoate
hydrochloride synthesized in Reference Example 1-(c), and
0.977 ml of triethylamine in dichloromethane (20 ml),
1.36 g of (tert-butoxycarbonyl)glycine and 1.61 g of 1
(3-dimethylaminopropyl)-3-ethylcarbodiimide hydrochloride
were added at room temperature under a stream of argon,
221102_G2572WO English Translation (Final) clean copy
and the mixture was stirred at room temperature for 30
minutes.
After the completion of reaction, a saturated
aqueous solution of ammonium chloride was added to the
reaction mixture, and the mixed solution was subjected to
extraction with dichloromethane. The organic layer was
washed with saturated saline, dried over anhydrous sodium
sulfate, filtered, and concentrated under reduced
pressure. The concentration residue was purified by
medium-pressure preparative chromatography (silica gel,
eluting solvent: hexane:ethyl acetate) to obtain 2.92 g
of the title compound as a colorless oil.
Mass spectrum (ESI, m/z): 439 [M+H]+.
[0235] Reference Example 2-(d), etc.:
Corresponding starting compounds were treated in the
same manner as in Reference Example 1-(d) to obtain
compounds described in the following Tables 4-1 to 4-8.
221102_G2572WO English Translation (Final) clean copy
[ 02 3 6]
Table 4-1 Reference Structural formula Mass spectrum
0
Reference 0 N N Example 2-(d) H 0 (Cmz):483,485[M+H]+
Br CF 3
0 H Reference O N O ( m 5[ Example 3-(d) H 0 (ESI,mlz):531[M+H]
CF 3
0 H Reference O N m 4[+ Example 4-(d) H 0 (ESI,mlz):419[M+H
CF 3
0H Reference o ^N 04 E,/N M Example 5-(d) H o (ESI,mlz):435[M+H]
0 CF3
H Reference 0 1 N 'YN OJ 0>1 Example 6-(d) H 0 0 (ESI, m/z): 423 [M+H]+
F CF 3
H 1 Reference 0' N -- N r0M Example 7-(d) H 0 (ESI, mlz): 473 [M+H]+
F 3C CF 3
221102_G2572WO English Translation (Final) clean copy
[0237]
Table 4-2 Reference Example Structural formula Mass spectrum
Reference Example 0 N C02Me (ESI, m/z): 445 [M+Na]+ 9-(d) H 0 F
CF 3
Reference Example 0 N CO2Me 10-(d) H C ( mz):461[M+Na]
CF 3
Reference Example 0 N CO 2 Me (ESI, m/z): 445 [M+Na]+ 11-(d) 0 F
F 3C
Reference Example o N CO 2Ms (ESI, m/z): 461 [M+Na]+ 12-(d) H Nc
F 3C
0 H 0 N Reference Example H 0 (ESI,mlz):423[M+H]+ 13-(d) CF 3 F
0 H0 0 N N ON Reference Example H 14-(d) 0 0 (ESI, m/z): 419 [M+H]+
CF 3
221102_G2572WO English Translation (Final) clean copy
[ 02 3 8]
Table 4-3 Reference Example Structural formula Mass spectrum
O N N O Reference Example H 0 0 (ESI, m/z): 457 [M+H]+ 15-(f) CI CF3 F
O N Reference Example H ESI,mlz):445,447[M+H]+ 16-(d)0 0 EI /)44,47[Hj BrN OMe
Reference Example ON Om 7 17-(d) H (ESI, m/z): 427 [M+Na]+
C Cl
J H Reference Example 04 DUS m/z):N M 18-(d) H 0 0 (ESI, mlz): 449, 451 [M+H+
Br Cl
0 H
Referenc Example > OAN' N 0 (DUIS, mlz): 493 [M-H]
Br Br
Reference Example AN N 0- (ESI, m/z): 541, 543 [M+H]+ 20-(d) H 0
Br '
221102_G2572WO English Translation (Final) clean copy
[ 02 39]
Table 4-4 Reference Example Structural formula Mass spectrum
Reference Example o NI (ESI m 47MH 21-(f) H o (ESI,mlz):497[M+H]
CI I
Reference Example O N CO 2 Me (ESI, m/z): 389 [M+H]+ 22-(d) H oF
Cl
ReferenceExample Q N CO7Me (ESI, m/z): 433, 435 [M+H]+
Br
_J H Reference Example No N m 41MH Re H (ESI, m/z): 471 [M+H]+
F J-0 CF:3
25-(e) F 0 F 0 Reference Example : 0 NJ ClOF HN 1 25-e)H F (ESI, mlz): 437 [M+H]+
Ci 0 ',F
Reference Example N- ESI, mz): 481,483[M+H]+ 26-(e) H 0 F
Br 0 F
221102_G2572WO English Translation (Final) clean copy
[0240]
Table 4-5 Reference Example Structural formula Mass spectrum
0 N Reference Example H N 0 ESI, m/z): 421 [M+H]+ 27-(e) F
F F
Reference Example 0 NCOMe (ESI,mlz):421[M+H]+ 28-(e) H 0 F
F 0 Refeenc Example ESI, m/z): 469 [M+H]+
Reference Example 0 ESI /) N4 Reference Examp (ESI, m/z): 469 [M+H]+
F 1 NOF
0 H Reference Example o0 cN (ESNm 31-(d) 32-(d) HF (ESI,mlz):455[M+H]+
CO OCF 3
Reference Example Oj-1N 04 N 32-(d) H 0 0 (ESI, mlz): 499, 501 [M+H]+
Br 0CF 3
0 H )NO AlN-, N 0 e Reference Example H 0 N CO 2 E Smz:49[H] 33-(d)F(EImz43[ H] F 3C0
221102_G2572WO English Translation (Final) clean copy
[0241]
Table 4-6 Reference Example Structural formula Mass spectrum
ON 5H 0 N Reference Example H 0 O (ESI,mlz):439[M+H]+ 34-(d) 0(Smz:49[+] OCF3 F
Reference Example ONESI, m 405 [M+H]+ 35-(d) H 0 (ESI, m/z): 405 [M+H]+
CF 3
F ( m 4 M Reference Example 36-(d) H F (ESI, m/z): 443 [M+Na]+
QCF 3
0 H Reference Example (ESI, m/z): 403 [M+H]+
0 149 F
O0 N N ,
Reference Example H 0 0 (ESI, mlz): 409 [M+Na]+ 38-(d)I
F
ON H Reference Example H 0 0 (ESI, mlz): 377 [M+H]+ 39-(d)
221102_G2572WO English Translation (Final) clean copy
[0242]
Table 4-7 Reference Example Structural formula Mass spectrum
Reference Example O K N O ESI, mlz):393 [M+H]+
Reference Example Q N ESI, mlz): 371 [M+H]+ 43-(d) H
ON Reference Example H- N (ESI, m/z): 403 [M+H]+
ReferenceExample N N Br Reerc EH (ESI, m/z): 431 [M+H]+
44 -(d)00
O N Reference Example H 0 0 (ESI, m/z): 43 [M+H]+ 45-(d) N
>O S1 0 N Reference Example H 0(Smz:42[+] N 45-(d)(EImz42[ H]
0
221102_G2572WO English Translation (Final) clean copy
[0243]
Table 4-8 Reference Example Structural formula Mass spectrum
Reference Example ( 0ESI, m/z): 425 [M+Na]+
O ,V N -XN 0 F O0
46-(d) EI /)42 M N]
Reference Example H 0o(ESI,m/z):427[M+Na]+
CF3
[0244] Reference Example l-(e)
Production of methyl (S)-3-(2-aminoacetamido)-3-(3
chloro-5-(trifluoromethyl)phenyl)propanoate hydrochloride
HCI
H 2N 0 0
CI CF 3
[0245] To a solution of 27.8 g of methyl (S)-3-(2-((tert
butoxycarbonyl)amino)acetamido)-3-(3-chloro-5
(trifluoromethyl)phenyl)propanoate synthesized in the
same manner as in Reference Example 1-(d) in cyclopentyl
methyl ether (190 ml), 60 ml of a 4 M solution of
hydrogen chloride in cyclopentyl methyl ether was added
at room temperature under a stream of argon, and the
mixture was stirred at room temperature for 7 hours.
221102_G2572WO English Translation (Final) clean copy
After the completion of reaction, the reaction
mixture was concentrated under reduced pressure to obtain
21.5 g of the title compound as a pale yellow oil.
Mass spectrum (ESI, m/z): 339 [M+H]+.
[0246] Reference Example 2-(e), etc.:
Corresponding starting compounds were treated in the
same manner as in Reference Example 1-(e) to obtain
compounds described in the following Tables 5-1 to 5-8.
221102_G2572WO English Translation (Final) clean copy
[0247]
Table 5-1 Reference Structural formula Mass spectrum
HCI H Reference H 2N (Cl, m/z): 383, 385 [M+H]+ Example 2-(e) 0
Br CF 3
HCI H
Reference H 2N N Os (ESI, m/z): 431 [M+H]+ Example 3-(e) 0 0
11 CF 3
HCI H N 1 Reference H 2N N OT Example 4-(e) 0 0 (ESI, m/z): 319 [M+H]+
CF 3
HCI H Reference H 2N N O S Example 5-(e) O 0 (ESI,mz):335[M+H]+
0 CF 3
HCI H Reference H 2N N O (ESI, m/z): 323 [M+H]+ Example 6-(e) 0 o
F CF3
HCI H
Reference H 2N N Os Example 7-(e) a0 (ESI, m/z): 373 [M+H]+ F 3C CF 3
221102_G2572WO English Translation (Final) clean copy
[0248]
Table 5-2 Reference Example Structural formula Mass spectrum HCI H H2 N N 0s R rence Example (ESI, m/z): 371 [M+H]+
F 3C N
HCI H Reference Example H 2N N CO 2Me (ESI, m/z): 323 [M+H]+ 9-(e) 0 F(EImz32[H] CF 3
HCI H Reference Example H 2N N 2Me (ESI, m/z): 339 [M+H]+ 1O0-(e) 0 Cl (EImz39[H]
CF 3
HCI H Reference Example H2N N CO 2Me (ESI, m/z): 323 [M+H]+ 1-(e) 0 F(EImz32[H]
HCI H Reference Example H 2N ESI, m/z): 33 [M+H]+ 12-(e) 0 Cl (EImz39[H]
F 3 CN
HCI H 0 (ES--mlz):323[M+H] Reference Example 13-(e) N
CF 3 F
221102_G2572WO English Translation (Final) clean copy
[0249]
Table 5-3 Reference Example Structural formula Mass spectrum HCI H H2N N O Reference Example (0 ESI, m/z): 357 [M+H]+ 15-(g)I CI CF 3 F HCI H H 2N N O Reference Example 0 0 (ESI, m/z): 345, 347 [M+H]+ 16-(e) Br OMe
HCI H Reference Example H 2N (ESI, m/z): 305 [M+H]+ 17-(e) ( 3
CI Cl
HCI H Reference Example H2N N O N(ESI, m/z): 349,351 [M+H]+ 18-(e) 0 0 EImz:4,5[+]
Br Cl
HCI H Reference Example H 2N (ESI, m/z): 395 [M+H]+ 19-(e) O O
Br Br
HCI H Reference Example H 2N N (ESI, m/z): 441, 443 [M+H]+ 20-(e) O O
Br I
221102_G2572WO English Translation (Final) clean copy
[0250]
Table 5-4 Reference Example Structural formula Mass spectrum
HCI H Reference Example H 2N N (ESI, m/z): 397 [M+H]+ 21-(g) 0 P
CI I
HCI H Reference Example H2N N CO 2 Me (ESI, m/z): 289 [M+H]+ 22-(e) 0 F
CI
HCI H Reference Example H 2N N CO 2 Me (ESI, m/z): 333, 335 [M+H]+ 23-(e) 0 F(EImz33,5M+f
Br
HCI H Reference Example H 2N (ESI, m/z): 371 [M+H]+ 24-(f) F 0 O
F 0 CF 3
HCI H Reference Example H2N N CO1 (ESI, m/z): 337 [M+H]+ 25-(f) 0 F (EImz37[ H] Cl 0 'jF
HCI H Reference Example H2N CO 2 Me (ESI, m/z): 381, 383 [M+H]+ 26-(f) 0 F
Br 0 F
221102_G2572WO English Translation (Final) clean copy
[ 0 2 51]
Table 5-5 Reference Example Structural formula Mass spectrum HCI H H2 N N CO 2 Me Reference Example O F (ESI, m/z): 321 [M+H]+ 27-(f) F
F
HCI H Reference Example H 2N N CO 2 Me (ESI, m/z): 321 [M+H]+ 28-(f) IF 0 . F(EImz32MH]
F 0 HCI H H2 N N CO 2 Me Reference Example 0 29-(b) F (ESI, m/z): 369 [M+H]+ F 0 N N
HCI H Reference Example H2N N Os (ESI, m/z): 369 [M+H]+ 30-(g) F O O F
F 0 O F
HCI H Reference Example H 2N N 0s (ESI, m/z): 355 [M+H]+ 31 -(e) 0 0 ' '
CI OCF 3
HCI H Reference Example H 2N N 0 N (ESI, m/z): 399,401 [M+H]+ 32-(e) 0 0
Br OCF 3
221102_G2572WO English Translation (Final) clean copy
[0252]
Table 5-6 Reference Example Structural formula Mass spectrum
HCI H
Reference Example H2N N CO 2 Me 33-(e) 0 F (ESI, m/z): 339 [M+H]+
F 3CO
HCI H H 2N N 0s Reference Example (O ESI, m/z): 339 [M+H]+ 34-(e)I OCF 3 F
HCI H Reference Example H 2N N Os (ESI, m/z): 305 [M+H]+ 35-(e) (E0ml)35[0]
CF 3
HCI H Reference Example H2N (ESI, m/z): 321 [M+H]+ 36-(e) 0 0
OCF 3
HCI H Reference Example H2 N N Os (ESI, m/z): 303 [M+H]+ 37-(e) O O F
0 F HCI H H 2N N Os Reference Example 0 0 (ESI, m/z): 287 [M+H]+ 38-(e) - 15 F
221102_G2572WO English Translation (Final) clean copy
[ 02 5 3]
Table 5-7 Reference Example Structural formula Mass spectrum HCI H
Reference Example H2N NS ) [ 39-(e) 0 6 0 (ESI, mlz): 277 [M+H]+
HCI H Reference Example H 2N N (ESI, m/z): 293 [M+H]+ 40-(e) ( 2 1 0
HCI H Reference Example H 2N N O (ESI, m/z): 271 [M+H]+ 41 -(e) (E0ml)210+]
CI
HCI H Reference Example H 2N N O (ESI, m/z): 315, 317 [M+H]+ 42-(e) 0 (E0mz:1,1[+]
Br
HCI H H-2N -- yN O Reerne xap ReferenceExample 0 0 (ESI, m/z): 303 [M+H]+ 43-(e) N'N
HCI H H 2N N O Reference Example 0 0 (ESI, m/z): 331 [M+H]+ 44-(e) -N
221102_G2572WO English Translation (Final) clean copy
[0254]
Table 5-8 Reference Example Structural formula Mass spectrum HCI H H2N N Reference Example 0 0 (ESI, m/z): 322 [M+H]+ 45-(e) N 0 HCI H H2N N O Reference Example (ESI, m/z): 303 [M+H]+ 46-(e) 0 F F
HCI H H2 N O ¾N Reference Example 0 0 (ESI, m/z): 305 [M+H]+ 47-(e)I
CF 3
[0255] Reference Example 18-(c)
Production of methyl (S)-3-amino-3-(3-bromo-5
chlorophenyl)propanoate hydrochloride
HCI H 2N 0
Br C
[0256] To a solution of 2.26 g of methyl (S)-3-(3-bromo
-chlorophenyl)-3-(((R)-tert
butylsulfinyl)amino)propanoate synthesized in Reference
Example 18-(b) in 1,4-dioxane (20 ml), 0.23 ml of
methanol and 4.1 ml of a 4 M solution of hydrogen
221102_G2572WO English Translation (Final) clean copy
chloride in 1,4-dioxane were added at room temperature
under a stream of argon, and the mixture was stirred at
room temperature for 18 hours.
After the completion of reaction, 60 ml of hexane
was added to the reaction mixture, and the mixture was
stirred at room temperature for 1 hour. The reaction
mixture was concentrated under reduced pressure. 10 ml
of tert-butyl methyl ether was added to the concentration
residue, and the mixture was stirred at room temperature
for 3 hours. A solid was collected by filtration and
dried under reduced pressure to obtain 1.07 g of the
title compound as a white solid.
Mass spectrum (ESI, m/z): 292, 294 [M+H]+.
[0257] Reference Example 7-(c), etc.:
Corresponding starting compounds were treated in the
same manner as in Reference Example 18-(c) to obtain
compounds described in the following Table 6.
221102_G2572WO English Translation (Final) clean copy
[ 02 5 8]
Table 6 Reference Example Structural formula Mass spectrum
HCI
Reference Example H 2N Os 7-(c)
F3 C CF 3
HCI
H2 N 0 ReferenceExample 14-(c) (ESI, m/z): 262 [M+H+
CF 3
HCI H2N O0 Reference Example 34-(c) 0 OCF 3 F
HCI
Reference Example H2N 0 (ESI, m/z): 214 [M+H]+ 41-(c) (
CI HCI
Reference Example H 2N 0s (ESI, m/z): 258, 260 [M+H]+ 42-(c) (E0mz:5,6[+] Br
[0259] Reference Example 8-(a)
Production of methyl (S)-3-(3-(lH-pyrazol-1-yl)-5
(trifluoromethyl)phenyl)-3-(2-((tert
butoxycarbonyl)amino)acetamido)propanoate
221102_G2572WO English Translation (Final) clean copy
J, HY 0 Fa 0
F 3C N -N
[0260] To a solution of 300 mg of methyl (S)-3-(2-((tert
butoxycarbonyl)amino)acetamido)-3-(3-bromo-5
(trifluoromethyl)phenyl)propanoate synthesized in the
same manner as in Reference Example 2-(d) in N,N
dimethylformamide (3 ml), 51 mg of pyrazole, 392 il of
trans-N,N'-dimethylcyclohexane-1,2-diamine, and 345 mg of
potassium carbonate were added at room temperature under
a stream of argon, and the mixture was stirred at room
temperature for 10 minutes under argon bubbling.
Subsequently, 236 mg of copper(I) iodide was added
thereto at room temperature, and the mixture was stirred
at 900C for 3 hours.
After the completion of reaction, water was added to
the reaction mixture, and the mixed solution was
subjected to extraction with ethyl acetate. The organic
layer was washed with a saturated aqueous solution of
ammonium chloride and saturated saline, dried over
anhydrous magnesium sulfate, filtered, and concentrated
under reduced pressure. The obtained residue was
purified by medium-pressure preparative chromatography
(silica gel, eluting solvent: hexane:ethyl acetate) to
obtain 81 mg of the title compound as a colorless oil.
Mass spectrum (ESI, m/z): 471 [M+H]+.
221102_G2572WO English Translation (Final) clean copy
[0261] Reference Example 29-(a):
A corresponding starting compound was treated in the
same manner as in Reference Example 8-(a) to obtain a
compound described in the following Table 7.
[0262]
Table 7 Reference Example Structural formula Mass spectrum
Reference Example N (ESI, m/z): 469 [M+H]+ 29-(a) F
[0263] Reference Example 14-(e)
Production of methyl (S)-3-(2-aminoacetamido)-3-(4
methyl-3-(trifluoromethyl)phenyl)propanoate hydrochloride
Hc H
0 0
CF
[0264] To 293 mg of methyl (S)-3-(2-((tert
butoxycarbonyl)amino)acetamido)-3-(4-methyl-3
(trifluoromethyl)phenyl)propanoate synthesized in
Reference Example 14-(d), 3.2 ml of tert-butyl methyl
ether and 0.657 ml of a 4 M solution of hydrogen chloride
in 1,4-dioxane were added in a nitrogen atmosphere, and
the mixture was stirred at room temperature for 3 hours.
0.657 ml of a 4 M solution of hydrogen chloride in 1,4
dioxane was further added thereto, and the mixture was
221102_G2572WO English Translation (Final) clean copy
stirred at room temperature for 3 hours. 0.657 ml of a 4
M solution of hydrogen chloride in 1,4-dioxane was
further added thereto, and the mixture was stirred at
room temperature for 2 hours. After the completion of
reaction, the reaction mixture was concentrated under
reduced pressure to obtain 259 mg of the title compound.
Mass spectrum (ESI, m/z): 319 [M+H]+.
[0265] Reference Example 15-(a)
Production of (3-chloro-4-fluoro-5
(trifluoromethyl)phenyl)methanol
CI' ' CF
[0266] To a solution of 1.00 g of 3-chloro-4-fluoro-5
(trifluoromethyl)benzoic acid in tetrahydrofuran (20 ml),
9.16 ml of a 0.9 M solution of a borane-tetrahydrofuran
complex in tetrahydrofuran was added in an ice bath under
a stream of argon, and the mixture was stirred at room
temperature for 20 hours.
After the completion of reaction, 2 M hydrochloric
acid was added to the reaction mixture, and the mixed
solution was subjected to extraction with ethyl acetate.
The organic layer was washed with a saturated aqueous
solution of sodium bicarbonate, dried over anhydrous
magnesium sulfate, filtered, and concentrated under
reduced pressure. The obtained residue was purified by
221102_G2572WO English Translation (Final) clean copy
medium-pressure preparative chromatography (silica gel,
eluting solvent: hexane:ethyl acetate) to obtain 1.06 g
of the title compound as a colorless oil.
Mass spectrum (EI, m/z): 228 [M]+.
[0267] Reference Example 21-(a), etc.:
Corresponding starting compounds were treated in the
same manner as in Reference Example 15-(a) to obtain
compounds described in the following Table 8.
[0268]
Table 8 Reference Example Structural formula Mass spectrum
HO Reference Example CEl, m/z): 268 [M]+ 21 -(a) I
HO Reference Example El, m/z): 240 [M]+ 30-(a) F F F O 0 F
[0269] Reference Example 21-(b)
Production of 3-chloro-5-iodobenzaldehyde
Os
[0270] To a solution of 2.04 g of (3-chloro-5
iodophenyl)methanol synthesized in Reference Example 21
(a) in dichloromethane (40 ml), 4.83 g of Dess-Martin
periodinane was added at room temperature with stirring
221102_G2572WO English Translation (Final) clean copy
under a stream of argon, and the mixture was stirred at
room temperature for 10 minutes.
After the completion of reaction, a saturated
aqueous solution of sodium bicarbonate supplemented with
sodium thiosulfate was added to the reaction solution,
and the mixed solution was subjected to extraction with
methylene chloride. The organic layer was washed with
saturated saline, dried over anhydrous magnesium sulfate,
filtered, and concentrated under reduced pressure. The
obtained residue was purified by medium-pressure
preparative chromatography (silica gel, eluting solvent:
hexane:ethyl acetate) to obtain 1.66 g of the title
compound as a colorless oil.
Mass spectrum (EI, m/z): 266 [M]+.
[0271] Reference Example 15-(b), etc.:
Corresponding starting compounds were treated in the
same manner as in Reference Example 21-(b) to obtain
compounds described in the following Table 9.
[0272]
Table 9 Reference Example Structural formula Mass spectrum 0
Reference Example (Cl, m/z): 227 [M+H]+ 15-(b) I C1 CF3 F
0 Reference Example 30-(b) F F F 0 0 F
221102_G2572WO English Translation (Final) clean copy
[0273] Reference Example 24-(a)
Production of 3-(difluoromethoxy)-5
(trifluoromethyl)benzaldehyde
0
F -O CF3
[0274] To a suspension of 4.36 g of potassium carbonate
in N,N-dimethylformamide (16 ml), a solution of 6.42 g of
sodium chlorodifluoroacetate and 4.00 g of 3-hydroxy-5
(trifluoromethyl)benzaldehyde in N,N-dimethylformamide
(20 ml) was added dropwise at 95°C under a stream of
argon, and the mixture was stirred at 95°C for 30
minutes.
After the completion of reaction, water was added to
the reaction mixture, and the mixed solution was
subjected to extraction with tert-butyl methyl ether.
The organic layer was washed with saturated saline, dried
over anhydrous magnesium sulfate, filtered, and
concentrated under reduced pressure. The obtained
residue was purified by medium-pressure preparative
chromatography (silica gel, eluting solvent: hexane:ethyl
acetate) to obtain 1.48 g of the title compound as a
colorless oil.
Mass spectrum (EI, m/z): 240 [M]+.
[0275] Reference Example 25-(a), etc.:
221102_G2572WO English Translation (Final) clean copy
Corresponding starting compounds were treated in the
same manner as in Reference Example 24-(a) to obtain
compounds described in the following Table 10.
[027 6]
Table 10 Reference Example Structural formula Mass spectrum
0 Reference Example (El, m/z): 206 [M]+ 25-(a) F (lmz:0[] Cl 0) F O Reference Example (El, m/z): 250, 252 [M]+ 26-(a) ~' F (lmz:5,5[ Br 0 F
Reference Example O F (El, m/z): 190 [M]+ 27-(a)0 F
F
O Reference Example F (El, m/z): 190 [M]+ 28-(a) F F (lmz:9[] F 0
[0277] Reference Example 1-(f)
Production of 2-fluoromalonamide
O 0
H2N NH2
[0278]
To a solution of 25.0 g of diethyl 2-fluoromalonate
in methanol (100 ml), 80 ml of a 7 N solution of ammonia
in methanol was added at room temperature under a stream
221102_G2572WO English Translation (Final) clean copy
of argon, and the mixture was stirred at room temperature
for 4 hours.
After the completion of reaction, a solid was
collected by filtration, washed with methanol, and then
dried under reduced pressure at 500C to obtain 15.91 g of
the title compound as a white solid.
'H-NMR spectrum (400MHz, DMSO-d 6 ) 6: 7.78 - 7.48 (m,
4H), 5.16 (d, J = 48.9 Hz, 1H).
[0279] Reference Example 1-(g)
Production of 2-fluoropropane-1,3-diamine hydrochloride
HCI
H 2N NH 2 F
[0280] To 400 ml of a 0.9 M solution of a borane
tetrahydrofuran complex in tetrahydrofuran, 7.20 g of 2
fluoromalonamide synthesized in Reference Example 1-(f)
was added in divided portions at room temperature under a
stream of argon, and the mixture was stirred at room
temperature for 20 hours.
After the completion of reaction, the reaction
mixture was cooled to 0°C, and 73 ml of methanol was
added dropwise thereto at 20°C or lower. The mixture was
stirred at room temperature for 1 hour and concentrated
under reduced pressure. 40 ml of ethanol was added to
the concentration residue, then 60 ml of a 2 M solution
of hydrogen chloride in ethanol was added thereto at 0°C,
and the mixture was stirred at room temperature for 3
221102_G2572WO English Translation (Final) clean copy
hours. A deposited solid was collected by filtration,
and the solid was dissolved in methanol and concentrated
under reduced pressure. To the obtained solution of 6.91
g of the solid in methanol (200 ml), 70 g of Amberlite
(IRA478RF CL) sequentially washed with 200 ml of water
and 200 ml of methanol was added at room temperature, and
the mixture was stirred at room temperature for 30
minutes. The reaction mixture was filtered, and the
filtrate was concentrated under reduced pressure to
obtain 4.55 g of the title compound as a white solid.
'H-NMR spectrum (400MHz, DMSO-d 6 +D 20) 6: 4.77 - 4.53
(m, 1H), 3.01 - 2.79 (m, 4H) .
[0281] Reference Example 48-(a)
Production of methyl 4-bromo-3-fluoro-1-(tetrahydropyran
2-yl)-1H-indazole-6-carboxylate
0
N-N
0
[0282] To a suspension of 200 mg of methyl 4-bromo-1H
indazole-6-carboxylate in acetonitrile (8 ml), 333 mg of
1-chloromethyl-4-fluoro-1,4-diazoniabicyclo[2.2.2]octane
bis(tetrafluoroborate) was added at room temperature
under a stream of argon, and the mixture was stirred at
1000C for 4 hours.
After the completion of reaction, water was added to
the reaction mixture, and the mixed solution was
221102_G2572WO English Translation (Final) clean copy
subjected to extraction with ethyl acetate. The organic
layer was washed with saturated saline, dried over
anhydrous sodium sulfate, filtered, and concentrated
under reduced pressure.
To a suspension of the obtained concentration
residue in dichloromethane (8 ml), 143 pl of 3,4-dihydro
2H-pyran and 15 mg of p-toluenesulfonic acid monohydrate
were added at room temperature under a stream of argon,
and the mixture was stirred at room temperature for 4
hours.
After the completion of reaction, a saturated
aqueous solution of sodium bicarbonate was added to the
reaction mixture, and the mixed solution was subjected to
extraction with dichloromethane. The organic layer was
washed with water, dried over anhydrous magnesium
sulfate, filtered, and concentrated under reduced
pressure. The obtained residue was subjected to medium
pressure preparative chromatography (silica gel, eluting
solvent: hexane:ethyl acetate), and a fraction containing
the compound of interest was concentrated under reduced
pressure.
The obtained residue was purified by gel permeation
chromatography (column: YMC-GPC T30000, YMC-GPC T4000,
and YMC-GPC T2000, eluting solvent: ethyl acetate) to
obtain 32 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 357, 359 [M+H]+.
[0283] Reference Example 49-(a)
221102_G2572WO English Translation (Final) clean copy
Production of methyl 3-chloro-4-nitro-1H-indazole-6
carboxylate
0 2N
ClO N-NHi
[0284] To a suspension of 300 mg of methyl 4-nitro-1H
indazole-6-carboxylate in acetonitrile (8 ml), 217 mg of
N-chlorosuccinimide was added at room temperature under a
stream of argon, and the mixture was stirred at 800C for
2 hours. Next, 145 mg of N-chlorosuccinimide was further
added thereto at 80°C, and the mixture was stirred at
°C for 2 hours.
After the completion of reaction, the reaction
mixture was poured to a saturated aqueous solution of
sodium bicarbonate, and the mixed solution was subjected
to extraction with ethyl acetate. The organic layer was
washed with saturated saline, dried over anhydrous
magnesium sulfate, filtered, and concentrated under
reduced pressure to obtain 347 mg of the title compound
as a pale yellow solid.
Mass spectrum (ESI, m/z): 254 [M-H]-.
[0285] Reference Example 1-(h)
Production of methyl 4-bromo-l-(tetrahydropyran-2-yl)-1H
indazole-6-carboxylate
221102_G2572WO English Translation (Final) clean copy
0
[0286] To a suspension of 1.00 g of methyl 4-bromo-1H
indazole-6-carboxylate in dichloromethane (10 ml), 717 pl
of 3,4-dihydro-2H-pyran and 82 mg of p-toluenesulfonic
acid monohydrate were added at room temperature under a
stream of argon, and the mixture was stirred at room
temperature for 1 hour.
After the completion of reaction, a saturated
aqueous solution of sodium bicarbonate was added to the
reaction mixture, and the mixed solution was subjected to
extraction with dichloromethane. The organic layer was
washed with water, dried over anhydrous magnesium
sulfate, filtered, and concentrated under reduced
pressure. The obtained residue was purified by medium
pressure preparative chromatography (silica gel, eluting
solvent: hexane:ethyl acetate) to obtain 1.13 g of the
title compound as a pale yellow solid.
Mass spectrum (ESI, m/z): 339, 341 [M+H]+.
[0287] Reference Example 49-(b):
A corresponding starting compound was treated in the
same manner as in Reference Example 1-(h) to obtain a
compound described in the following Table 11.
221102_G2572WO English Translation (Final) clean copy
[ 02 8 8]
Table 11 Reference Example Structural formula Mass spectrum 0
Reernc Eamle 02 N Reference4Example c -1-0 (ESI, m/z): 340 [M+H]+ N-N
[0289] Reference Example 1-(i)
Production of methyl 4-((diphenylmethylene)amino)-1
(tetrahydropyran-2-yl)-1H-indazole-6-carboxylate
N-N 0
[0290] To a solution of 1.13 g of methyl 4-bromo-l
(tetrahydropyran-2-yl)-1H-indazole-6-carboxylate
synthesized in Reference Example 1-(h) in xylene (20 ml),
1.11 ml of benzophenone imine, 305 mg of
tris(dibenzylideneacetone)dipalladium(0), 771 mg of 4,5
bis(diphenylphosphino)-9,9-dimethylxanthene, and 3.26 g
of cesium carbonate were added at room temperature under
a stream of argon. After purging with an argon
atmosphere, the mixture was stirred at 150°C for 6 hours
under a stream of argon.
After the completion of reaction, a saturated
aqueous solution of ammonium chloride was added to the
reaction mixture, and the mixed solution was subjected to
221102_G2572WO English Translation (Final) clean copy
extraction with toluene. The organic layer was washed
with saturated saline, dried over anhydrous sodium
sulfate, filtered, and concentrated under reduced
pressure. The obtained residue was subjected to medium
pressure preparative chromatography (silica gel, eluting
solvent: hexane:ethyl acetate), and a fraction containing
the compound of interest was concentrated under reduced
pressure. 20 mL of tert-butyl methyl ether was added to
the obtained residue at room temperature, and the mixture
was stirred at room temperature for 1 hour. A solid was
collected by filtration to obtain 1.09 g of the title
compound as a yellow solid.
'H-NMR spectrum (400MHz, DMSO-d6 ) 6: 8.09 (d, J = 0.8
Hz, 1H), 7.90 - 7.87 (m, 1H), 7.78 - 7.70 (m, 2H), 7.62
7.46 (m, 3H), 7.28 - 7.17 (m, 5H), 6.91 (d, J = 1.1 Hz,
1H), 5.91 (dd, J = 2.3, 9.5 Hz, 1H), 3.89 - 3.72 (m, 5H),
2.40 - 2.29 (m, 1H), 2.04 - 1.92 (m, 2H), 1.80 - 1.68 (m,
1H), 1.61 - 1.49 (m, 2H).
[0291] Reference Example 48-(b):
A corresponding starting compound was treated in the
same manner as in Reference Example 1-(i) to obtain a
compound described in the following Table 12.
221102_G2572WO English Translation (Final) clean copy
[0292]
Table 12 Reference Example Structural formula Mass spectrum Ph Ph
Reference Example F (ESI, m/z): 458 [M+H]+ 48-(b) F v(SXlz:5[+] N-N
[0293] Reference Example 1-(j)
Production of methyl 4-amino-i-(tetrahydropyran-2-yl)-lH
indazole-6-carboxylate
0 H2N
I-N
[0294] To a solution of 890 mg of methyl 4
((diphenylmethylene)amino)-1-(tetrahydropyran-2-yl)-1H
indazole-6-carboxylate synthesized in Reference Example
1-(i) in tetrahydrofuran (30 ml), 970 mg of 5% palladium
carbon (containing 50.57% water, AER-type manufactured by
N.E. Chemcat Corp.) was added at room temperature under a
stream of argon. After purging with a hydrogen
atmosphere, the mixture was stirred at room temperature
for 1.5 hours.
After the completion of reaction, the reaction
mixture was filtered through celite and washed with ethyl
acetate, and the filtrate was concentrated under reduced
pressure. The obtained residue was purified by medium
221102_G2572WO English Translation (Final) clean copy
pressure preparative chromatography (silica gel, eluting
solvent: hexane:ethyl acetate) to obtain 523 mg of the
title compound as a white solid.
Mass spectrum (ESI, m/z): 276 [M+H]+.
[0295] Reference Example 48-(c)
Production of methyl 4-amino-3-fluoro-1-(tetrahydropyran
2-yl)-1H-indazole-6-carboxylate
H2N
F N-N
0
[0296] To a solution of 100 mg of methyl 4
((diphenylmethylene)amino)-3-fluoro-1-(tetrahydropyran-2
yl)-1H-indazole-6-carboxylate synthesized in Reference
Example 48-(b) in tetrahydrofuran (0.4 ml), 0.8 ml of a
% aqueous citric acid solution was added at room
temperature under a stream of argon, and the mixture was
stirred at 700C for 18 hours.
After the completion of reaction, a saturated
aqueous solution of sodium bicarbonate was added to the
reaction mixture, and the mixed solution was subjected to
extraction with ethyl acetate. The organic layer was
washed with saturated saline, dried over anhydrous sodium
sulfate, filtered, and concentrated under reduced
pressure. The obtained residue was purified by medium
pressure preparative chromatography (silica gel, eluting
221102_G2572WO English Translation (Final) clean copy
solvent: hexane:ethyl acetate) to obtain 53 mg of the
title compound as a yellow solid.
Mass spectrum (ESI, m/z): 294 [M+H]+.
[0297] Reference Example 49-(c)
Production of methyl 4-amino-3-chloro-1-(tetrahydropyran
2-yl)-1H-indazole-6-carboxylate
0
N-N 0
[0298] To a suspension of 304 mg of methyl 3-chloro-4
nitro-1-(tetrahydropyran-2-yl)-1H-indazole-6-carboxylate
synthesized in Reference Example 49-(b) in ethanol (3 ml)
and water (1.5 ml), 779 mg of sodium dithionite was added
at room temperature under a stream of argon, and the
mixture was stirred at 900C for 1 hour.
After the completion of reaction, the reaction
mixture was poured to water, and the mixed solution was
subjected to extraction with ethyl acetate. The organic
layer was washed with a saturated aqueous solution of
sodium bicarbonate, dried over anhydrous magnesium
sulfate, filtered, and concentrated under reduced
pressure. The obtained residue was purified by medium
pressure preparative chromatography (silica gel, eluting
solvent: hexane:ethyl acetate) to obtain 131 mg of the
title compound as a pale yellow solid.
Mass spectrum (ESI, m/z): 310 [M+H]+.
221102_G2572WO English Translation (Final) clean copy
[0299] Reference Example 1-(k)
Production of methyl 4-(3-methylthioureido)-l
(tetrahydropyran-2-yl)-1H-indazole-6-carboxylate
P0 H
[0300] To a solution of 200 mg of methyl 4-amino-1
(tetrahydropyran-2-yl)-1H-indazole-6-carboxylate
synthesized in the same manner as in Reference Example 1
(j) in N-methylpyrrolidone (2 ml), 133 mg of methyl
isothiocyanate and 200 ptl of acetic acid were added at
room temperature under a stream of argon, and the mixture
was stirred at 1000C for 9 hours.
After the completion of reaction, water was added to
the reaction mixture, and the mixed solution was
subjected to extraction with ethyl acetate. The organic
layer was washed with saturated saline, dried over
anhydrous magnesium sulfate, filtered, and concentrated
under reduced pressure. The obtained residue was
purified by medium-pressure preparative chromatography
(silica gel, eluting solvent: hexane:ethyl acetate) to
obtain 184 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 349 [M+H]+.
[0301] Reference Example 48-(d), etc.:
221102_G2572WO English Translation (Final) clean copy
Corresponding starting compounds were treated in the
same manner as in Reference Example 1-(k) to obtain
compounds described in the following Table 13.
[0302]
Table 13 Reference Example Structural formula Mass spectrum
HN S
Reference Example HN (ESI, m/z): 367 [M+H]+ 48-(d) F(EIm )36[+] N-N
HN S
Reference Example cH o (ESI, m/z): 383
[M+H]+ 49-(d) ci(EIm )38[+] N-N
0
[0303] Reference Example 52-(a)
Production of methyl 6-(3-methylthioureido)-lH-indazole
4-carboxylate
H- H C C YN
[0304] To a solution of 500 mg of methyl 6-amino-lH
indazole-4-carboxylate in N-methylpyrrolidone (2.5 ml),
233 mg of methyl isothiocyanate was added at room
temperature under a stream of argon, and the mixture was
stirred at room temperature for 2 hours, left standing at
room temperature for 84 hours, and stirred at 70°C for 7
hours.
221102_G2572WO English Translation (Final) clean copy
After the completion of reaction, water was added to
the reaction mixture, and the mixture was stirred at room
temperature for 15 minutes. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 408 mg of
the title compound as a pale yellow solid.
Mass spectrum (ESI, m/z): 265 [M+H]+.
[0305] Reference Example 1-(l)
Production of 4-(3-methylthioureido)-1H-indazole-6
carboxylic acid
Y OH S W N-NH
[0306] To a suspension of 4.81 g of methyl 4-(3
methylthioureido)-1-(tetrahydropyran-2-yl)-1H-indazole-6
carboxylate synthesized in the same manner as in
Reference Example 1-(k) in methanol (69.2 ml), 17.3 ml of
a 4 M solution of hydrogen chloride in 1,4-dioxane was
added at room temperature under a stream of argon, and
the mixture was stirred at room temperature for 6 hours.
After the completion of reaction, the reaction
mixture was concentrated under reduced pressure,
subjected to azeotropy with 30 ml of tert-butyl methyl
ether, and dried under reduced pressure at 50°C.
To a suspension of the obtained residue in methanol
(41.4 ml), 41.4 ml of a 1 M aqueous sodium hydroxide
solution was added at room temperature under a stream of
221102_G2572WO English Translation (Final) clean copy
argon, and the mixture was stirred at room temperature
for 3 hours.
After the completion of reaction, the reaction
mixture was adjusted to pH 4.0 by the addition of 2 M
hydrochloric acid, and the mixture was stirred at room
temperature for 1 hour. A deposited solid was collected
by filtration, washed with water, and then dried under
reduced pressure at 60°C to obtain 2.80 g of the title
compound as a pale yellow solid.
Mass spectrum (ESI, m/z): 251 [M+H]+.
[0307] Reference Example 49-(e)
Production of methyl 3-chloro-4-(3-methylthioureido)-1H
indazole-6-carboxylate
HN S
C1 N-NH
[0308] To a suspension of 840 mg of methyl 3-chloro-4-(3
methylthioureido)-1-(tetrahydropyran-2-yl)-1H-indazole-6
carboxylate synthesized in the same manner as in
Reference Example 49-(d) in dichloromethane (5 ml), 2.52
ml of trifluoroacetic acid was added dropwise at room
temperature under a stream of argon, and the mixture was
stirred at room temperature for 10 hours.
After the completion of reaction, the reaction
mixture was concentrated under reduced pressure. The
obtained residue was purified by medium-pressure
221102_G2572WO English Translation (Final) clean copy
preparative chromatography (silica gel, eluting solvent:
hexane:ethyl acetate) to obtain 375 mg of the title
compound as a pale yellow solid.
Mass spectrum (ESI, m/z): 299 [M+H]+.
[0309] Reference Example 48-(e)
Production of 3-fluoro-4-(3-methylthioureido)-1
(tetrahydropyran-2-yl)-1H-indazole-6-carboxylic acid
kN5
HNH F N-N
[0310] To a suspension of 245 mg of methyl 3-fluoro-4-(3
methylthioureido)-1-(tetrahydropyran-2-yl)-1H-indazole-6
carboxylate synthesized in the same manner as in
Reference Example 48-(d) in methanol (2 ml), 2 ml of a 1
M aqueous sodium hydroxide solution and 3 ml of
tetrahydrofuran were added at room temperature under a
stream of argon, and the mixture was stirred at room
temperature for 2 hours.
After the completion of reaction, the reaction
mixture was adjusted to pH 4.0 by the addition of 1 M
hydrochloric acid, and the mixture was stirred at room
temperature for 1 hour. The reaction mixture was
concentrated under reduced pressure, and tetrahydrofuran
was distilled off under reduced pressure. The residue
was stirred at room temperature for 15 hours. A
deposited solid was collected by filtration, washed with
221102_G2572WO English Translation (Final) clean copy
water, and then dried under reduced pressure at 600C to
obtain 202 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 353 [M+H]+.
[0311] Reference Example 49-(f)
Production of 3-chloro-4-(3-methylthioureido)-1H
indazole-6-carboxylic acid
I HN S
HN ~OH CI N-NH
[0312] To a suspension of 337 mg of methyl 3-chloro-4-(3
methylthioureido)-1H-indazole-6-carboxylate synthesized
in Reference Example 49-(e) in methanol (4.2 ml), 1.69 ml
of a 2 M aqueous sodium hydroxide solution was added
dropwise at room temperature, and the mixture was stirred
at room temperature for 5 hours.
After the completion of reaction, the reaction
mixture was adjusted to pH 4 by the addition of 2 M
hydrochloric acid, and the mixture was stirred at room
temperature for 0.5 hours. A deposited solid was
collected by filtration and dried under reduced pressure
to obtain 110 mg of the title compound as a pale yellow
solid.
Mass spectrum (ESI, m/z): 285 [M+H]+.
[0313] Reference Example 52-(b):
221102_G2572WO English Translation (Final) clean copy
A corresponding starting compound was treated in the
same manner as in Reference Example 49-(f) to obtain a
compound described in the following Table 14.
[0314]
Table 14 Reference Example Structural formula Mass spectrum
H H O Reference Example -N N OH (ESI, m/z): 251 [M+H]+ 52-(b) S HN-N
[0315] Reference Example 1-(m)
Production of 4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2
yl)amino)-1H-indazole-6-carboxylic acid
HH
[0316] To a solution of 10.00 g of 4-(3
methylthioureido)-1H-indazole-6-carboxylic acid
synthesized in the same manner as in Reference Example 1
(1) in N-methylpyrrolidone (100 ml), 3.00 ml of
iodomethane was added at room temperature under a stream
of argon, and the mixture was stirred at room temperature
for 16 hours. The pressure of the reaction mixture was
reduced, and excess iodomethane was removed. 10.27 g of
2-fluoropropane-1,3-diamine hydrochloride synthesized in
the same manner as in Reference Example 1-(g) and 17 ml
of water were added to the obtained solution at room
temperature, and the mixture was stirred at 1100C for 8
221102_G2572WO English Translation (Final) clean copy
hours, stirred at room temperature for 13 hours, stirred
at 110°C for 11 hours, stirred at room temperature for 13
hours, and stirred at 110°C for 9 hours.
After the completion of reaction, 283 ml of water
was added to the reaction mixture, and the mixture was
stirred at room temperature for 15 hours. A deposited
solid was collected by filtration, washed with water, and
then dried under reduced pressure at 600C to obtain 7.40
g of the title compound as a white solid.
Mass spectrum (ESI, m/z): 278 [M+H]+.
[0317] Reference Example 48-(f), etc.:
Corresponding starting compounds were treated in the
same manner as in Reference Example 1-(m) to obtain
compounds described in the following Table 15.
[0318]
Table 15 Reference Example Structural formula Mass spectrum F
HN N O Reference Example HN OH (ESI, m/z): 380 [M+H]+ 48-(f)N N-N
F
Reference Example HN N O (ESI, m/z): 312 [M+H]+ 49-(g) HN OH CI N-NH
H H O Reference Example N N OH (ESI, m/z): 278 [M+H]+ 52-(c) F N
HN-N
221102_G2572WO English Translation (Final) clean copy
[0319] Reference Example 50-(a)
Production of 4-((5-hydroxy-1,4,5,6-tetrahydropyrimidin
2-yl)amino)-1H-indazole-6-carboxylic acid
H H H N N, OH NNH
[0320] To a solution of 1.00 g of 4-(3-methylthioureido)
1H-indazole-6-carboxylic acid synthesized in the same
manner as in Reference Example 1-(l) in N
methylpyrrolidone (10 ml), 0.30 ml of iodomethane was
added at room temperature under a stream of argon, and
the mixture was stirred at room temperature for 2 hours.
The pressure of the reaction mixture was reduced, and
excess iodomethane was removed. 1.08 g of 1,3
diaminopropan-2-ol was added to the obtained solution at
room temperature, and the mixture was stirred at 110°C
for 6 hours.
After the completion of reaction, the reaction
mixture was cooled to room temperature and stirred at
room temperature for 17 hours. 10 ml of water was added
thereto, and the mixture was stirred at room temperature
for 4 hours. A deposited solid was collected by
filtration, washed with water, and then dried under
reduced pressure to obtain 915 mg of the title compound
as a white solid.
Mass spectrum (ESI, m/z): 276 [M+H]+.
221102_G2572WO English Translation (Final) clean copy
[0321] Reference Example 51-(a):
A corresponding starting compound was treated in the
same manner as in Reference Example 50-(a) to obtain a
compound described in the following Table 16.
[0322]
Table 16 Reference Example Structural formula Mass spectrum
H H Reference Example N NOH (ESI,m/z):260[M+H]+ 51 -(a) NH (Smz:20[+l
N-NH
[0323] Reference Example 48-(g)
Production of 3-fluoro-4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6-carboxylic
acid F
HcI
F H F N-NH
[0324] To 110 mg of 3-fluoro-4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1-(tetrahydropyran-2-yl)
1H-indazole-6-carboxylic acid synthesized in the same
manner as in Reference Example 48-(f), 2.9 ml of 1 M
hydrochloric acid was added at room temperature under a
stream of argon, and the mixture was stirred at 60°C for
11 hours and stirred at room temperature for 13 hours.
Subsequently, 1.45 ml of 1 M hydrochloric acid was added
221102_G2572WO English Translation (Final) clean copy
thereto at room temperature, and the mixture was stirred
at 700C for 9 hours and stirred at room temperature for
18 hours.
After the completion of reaction, a solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 74 mg of
the title compound as a white solid.
Mass spectrum (ESI, m/z): 296 [M+H]+.
[0325] Example 1-(a)
Production of methyl (3S)-3-(3-chloro-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
0 H H 0 H N N N O0 F3C'OH F O N-NH CF 3
[0326] To a suspension of 80 mg of methyl (S)-3-(2
aminoacetamido)-3-(3-chloro-5
(trifluoromethyl)phenyl)propanoate hydrochloride
synthesized in the same manner as in Reference Example 1
(e) and 65 mg of 4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6-carboxylic
acid synthesized in the same manner as in Reference
Example 1-(m) in N,N-dimethylformamide (1 ml), 38 mg of
1-hydroxybenzotriazole was added at room temperature
under a stream of argon, and the mixture was stirred at
221102_G2572WO English Translation (Final) clean copy
room temperature for 10 minutes. Subsequently, 0.043 ml
of N,N'-diisopropylcarbodiimide was added thereto at room
temperature, and the mixture was stirred at room
temperature for 5 hours.
After the completion of reaction, the reaction
mixture was purified by medium-pressure preparative
chromatography (ODS, eluting solvent: 0.1% aqueous
trifluoroacetic acid solution:0.1% solution of
trifluoroacetic acid in acetonitrile) to obtain 132 mg of
the title compound as a pale yellow solid.
Mass spectrum (ESI, m/z): 598 [M+H]+.
[0327] Example 2-(a), etc.:
Corresponding starting compounds were treated in the
same manner as in Example 1-(a) to obtain compounds
described in the following Tables 17-1 to 17-7.
221102_G2572WO English Translation (Final) clean copy
[ 0 32 8]
Table 17-1
Example Structural formula Mass spectrum
H H B C Example F 3C OH NN O
N-Br CF 3 0 H H 0 H N-NHCF Example Example F3C OH F30HF NO NN (ESI, m/z): 5786 [M+H]+ OmN
3E(amp) F N O (ESI, m/z): 690 [M+H]+ H I CF 3
0 H H 0 H Example F 3C OH N N (S m/z) 62 [ Xap F N O (ESI, m/z): 578 [M+H]+ N-NH CF 3
0 H H 0 H Example F3C) OH N N N ' N 01 -(a) FU NF O F (ESI, m/z): 594 [M+H]+ N-NH 0 CF 3
H H192 Exml FkO N- N 0 FCC Fxml N H 0 (ESI, mlz): 582 [M+H]+ 6-(a) FC NH F CF 3
O H H 0 H Example F3 C" OH NN ANIY 7-() N0 0 (ESI, mlz): 632 [M+H]+ NH F3C CF 3 oH HwH
Example 3 ~H y H N 0 .- (ESI, mlz): 630 [M+H]+ 8-(a) F N-NH IC2 0N
O H H 0 H NIN N Example F3C OH Ij- H CO2 ESmz:52[H] 9-(a) F N0 F
NH CF 3
221102_G2572WO English Translation (Final) clean copy
[ 0 32 9 ]
Table 17-2
Example Structural formula Mass spectrum
0 H H N 0~ Example F3C OH F jN-' NH C0 2Me
+ Exame C F N (ESI, m/z): 598 [M+H]+ N-NH CF 3
0 H H O Example F3 C) O N NYN O2 M Examl F F N 0 FO (ESI, m/z): 582 [M+H]+ F3C
0 H H O Example 3F C OH f N N o ES FaC OF N N (ESI, m/z): 598 [M+H]+ N--NH CF3 PNH )'F3C
0 C H HN 0y ON
Example 3N OH I H p3(a) e F3C OH F O (ESI, m/z): 582 [M+H]+ CF 3
0 H H 0 H
F3C'OH N N N N 0 Example 14()FU N-N 0 0 (ESI, mlz): 578 [M+H]+ CF 3
Example IN OH HH0N
15-(a) F3CO N Io" (ESI, mlz): 616 [M+H]+ CI CF 3 F
0 H H 0 H
Example F3 C OH N' NN N 16()FU PH 0 1. 0 (ESI, mlz): 604,606 [M+H]+ 16-() N-H NBr
0 H H 0 H Exmpe 3 CkO ExmleFC NN N ~ O HC (ESI, mlz): 564 [M+H]+ N7-NH N 0 0
221102_G2572W0 English Translation (Final) clean copy
[ 033 0]
Table 17-3 Example Structural formula Mass spectrum
0 H H 0 H ExamplNF 3 CyO Ny0, 18-(a) F Nxml F3 O 0 0 (ESI, mlz): 608, 610 [M+H]+ N-HBr cI
0 H H 0 H Example F3C OH HEImz: 5N[+] 19-(a) F N 0 (E0mz:5[+] N-NH -!
NH Br Br
0 H H 0 H Example F3C OH H EI mz:70,0 [f] 20-(a) F N 0 0EImz:0,0[+] N-NH: NHBr I
0 H H 0 H Exaple F3 COH H (ESI, mlz): 656 [M+H]+ 21 -(a) F NNH ci I
O H H 0 H
NNH
0 H H 0 H Example F3C k OH NyNp N"'SN CO 2Me 23-(a F N 0 (ESI, mlz): 592, 594 [M+H]+ NNH Br
0 H H 0 H Example F3 c lOH N AN NI- N4(a F N 0 (ESI, mlz): 630 [M+H]+ NNH FO " CF 3
0 H H 0 H Example F3 C~ NHN (EI /) 56[+ 25-(a) F N 0 0. F (S~lz:9[+]
221102_G2572WO English Translation (Final) clean copy
[ 0 3 31]
Table 17-4
Example Structural formula Mass spectrum
0 H H 0 H Example F3CAOH N:N604[+ Ex-a)l FCO F O (ESI, m/z): 640,642[M+H] N-NH O F Br 0 F
Example F3C OH F (ES 6 Exam F N O (ESI, m/z): 614 [M+H]+ N-NH C OAF F
0 H H 0 H Example F3 H (E0 m/)H28[+] Example F3C OH N N (ESI,m/z):580[M+H]+ F H --- N N H F N N
28-(a) NH HF N-NH 0 F
0 H H 0 H Example F3C'kOH NyN - N 0 29-(a) F N\ 0 F (ESI, mlz): 628 [M+H]+ NH 0-- F ,
0 H H 0 H
30-(a) N N NNH N- )
Example F 3C OH FHN (ESI, m/z): 618 [M+H]+
Example F 3C OH NN N N? 0 (ESI, mlz):658,66[M+H] N 0- N2(a H H H195 BHrc OCF 3
0 H H 0 H F~&O N N N CO, Example F3 CH FCO CF +P
32-(a) 0 (ESI, mlz):58,0[M+H] FIU
NNH H
N-H F 3CO
221102_G2572WO English Translation (Final) clean copy
[0332]
Table 17-5
Example Structural formula Mass spectrum
H H N H
Example F3C OH F N N (ESI, m/z): 598 [M+H]+ N-NH N OCF 3 F
0 H H
Example F3C OH N N ?[N 35-(a) FIN H~ 0 0 (ESI, mlz): 564 [M+H]+ N-NH CF 3
0 H H Example F3C~ NP N-N rO E eO N N O (ESI, m/z): 580 [M+H]+ N-NH OCF 3
Example F3C 'OH F (EN m /zOM Ex-aml FCO F N (ESI, m/z): 562 [M+H]+ -NH C OH
0 H H 0 H
Example F3C OHF N (ESI,m/z):546[M+H]+
N-NH F. Example F3C OH F N IN ON (ESI, m/z): 536 [M+H]+ 3--(a) 16 0 H H 1 1 Example F 3 C OH NNP N N
39(a Z N(a Ff .- (ESI, mlz): 552[M+H]+
0 H H 0 H Example F 3C OH Ny N N~N 1 41(a F NH (ESI, mlz): 530[M+H]+ NNH N. "L
0~ 19 H -
221102_G2572WO English Translation (Final) clean copy
[ 0 3 3 3]
Table 17-6
Example Structural formula Mass spectrum
0 H H 0 H Example F3C OH F N N (ESI, m/z): 574,576 [M+H]+
N-NH Br
O H H
Example F3 C OH NyN OrN (5 FN 0 (ESI, m/z): 562 [M+H]+ 43-(a) F NH N
F3 C ) OH N0 Example F F N O (ESI,mz):590[M+H]+
O O
Example F3C OHF N O (ESI, m/z): 564 [M+H]+ N-NH N 0o 0 H
45-(a) \N-F NZi (ESI, mlz): 562[M+H]+ Example F3C OH H ON H- HI
N N N~y ON
Ex-a)pl F N0 0 0 (ESI, mlz): 564 [M+H]+ 46()N-NHN
Fr
Example F IH~y H 47-(a Ff i, W\0 1 0 (ESI, mlz):66[M+H]+ N-NH N CICF 3 H~j- 19 -~
F 9
221102_G2572WO English Translation (Final) clean copy
[0334]
Table 17-7
Example Structural formula Mass spectrum F
Examplo N, F3C)'OH H 0 H (ESI, m/z): 632 [M+H]+ N--)N 0 N-NH CI CF 3
O H H CF3 Example F3C OH HN OK (ESI, m/z): 596 [M+H]+ 50-(a) NO 0N
NH CF 3 O H H H Example F3C OH NN O ON (ESIm/z):58rM+H]+ HN- Cl CF3
N-N ci CF3
H H198 Example F3c OH NN ~ NH N-N g Br C c (ESI, mlz): 598 [M+H]+ 52-(a) F N/ HN-N 'N- CF
0 H H 0 H Example F 3C ) OH N N N N0, 'N
53-a) N -H (ESI, mlz): 642,644 [M+H]+ N-N C" HNN Br CF 3 Ex -a)pe FCO N N\ O0 (ESI, m/z): 564 [M+H]+ O H H 0 H Example 1 F3C ' "'OH N N N -N F NN4(a 0 (ESI, mlz): 690 [M+H]+ HNN I CF 3
O H H 0 H F~ N N N 0N Exmpe 3COH f j- H0 (EIm)58[+] Nxml N6Y 55-(a) F N 0 I E I /z:52[+ ] HN-N "NCF 3
0 H H 0 H Example F3C OH NN Ir F N6(a N0 (ESI, mlz): 564 [M+H]+ HN-N "NCF3
221102_G2572WO English Translation (Final) clean copy
[0335] Example 1-(b)
Production of (3S)-3-(3-chloro-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
H H O H N N N OH N N H o O F N-NH Cl C 3 I CF 3
[0336] To a suspension of 130 mg of methyl (3S)-3-(3
chloro-5-(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 1-(a) in acetonitrile (1.5 ml) and
water (1.5 ml), 22 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.2 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 2 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 58 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 584 [M+H]+.
221102_G2572WO English Translation (Final) clean copy
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 - 8.06 (m,
2H), 7.70 - 7.67 (m, 1H), 7.66 - 7.63 (m, 1H), 7.54
7.50 (m, 2H), 5.30 - 5.13 (m, 2H), 4.18 - 4.05 (m, 2H),
3.72 - 3.45 (m, 4H), 2.74 - 2.60 (m, 2H).
[0337] Example 2-(b)
Production of (3S)-3-(3-bromo-5-(trifluoromethyl)phenyl)
3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2
yl)amino)-1H-indazole-6-carboxamido)acetamido)propanoic
acid
H H H N N N INf CU 2 IF N-NH Br CF3
[0338] To a solution of 193 mg of methyl (3S)-3-(3-bromo
-(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 2-(a) in acetonitrile (3 ml) and
water (3 ml), 31 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 3 hours.
Subsequently, 36 mg of lithium hydroxide was further
added thereto at room temperature, and the mixture was
stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.8 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
221102_G2572WO English Translation (Final) clean copy
temperature for 16 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 60°C to obtain 107 mg of
the title compound as a white solid.
Mass spectrum (ESI, m/z): 628, 630 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.10 - 8.05 (m,
2H), 7.86 - 7.81 (m, 1H), 7.71 - 7.64 (m, 2H), 7.51 (d, J
= 1.1 Hz, 1H), 5.31 - 5.12 (m, 2H), 4.18 - 4.05 (m, 2H),
3.73 - 3.43 (m, 4H), 2.73 - 2.60 (m, 2H).
[0339] Example 3-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(3-iodo-5
(trifluoromethyl)phenyl)propanoic acid
H H 0 H N N N N OH
F N 0O
N-NH CF3
[0340] To a solution of 79 mg of methyl (3S)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(3-iodo-5
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 3-(a) in acetonitrile (1.5 ml) and
water (1.5 ml), 12 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1.5 hours.
221102_G2572WO English Translation (Final) clean copy
After the completion of reaction, the reaction
mixture was adjusted to pH 5.4 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 1 hour. A deposited solid was collected
by filtration, washed with water, and then dried under
reduced pressure at 600C to obtain 35 mg of the title
compound as a white solid.
Mass spectrum (ESI, m/z): 676 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 - 8.05 (m,
2H), 8.03 - 8.00 (m, 1H), 7.84 - 7.81 (m, 1H), 7.72
7.69 (m, 1H), 7.52 - 7.49 (m, 1H), 5.29 - 5.13 (m, 2H),
4.18 - 4.04 (m, 2H), 3.71 - 3.44 (m, 4H), 2.73 - 2.57 (m,
2H).
[0341] Example 4-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(3-methyl-5
(trifluoromethyl)phenyl)propanoic acid
H H O H N N N N OH
F N 0 0
N-NH CF 3
[0342] To a solution of 97 mg of methyl (3S)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(3-methyl-5
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 4-(a) in acetonitrile (1 ml) and
221102_G2572WO English Translation (Final) clean copy
water (1 ml), 17 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1.5 hours.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.8 by the addition of 1 N
hydrochloric acid, and the mixture was filtered. The
filtrate was stirred at room temperature for 17 hours,
and a deposited solid was collected by filtration, washed
with water, and then dried under reduced pressure at 600C
to obtain 46 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 564 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 (d, J = 0.9 Hz, 1H), 8.08 - 8.06 (m, 1H), 7.51 (d, J = 1.3 Hz, 1H),
7.49 - 7.45 (m, 2H), 7.33 - 7.30 (m, 1H), 5.30 - 5.14 (m,
2H), 4.19 - 4.04 (m, 2H), 3.73 - 3.45 (m, 4H), 2.75
2.63 (m, 2H), 2.39 (s, 3H).
[0343] Example 5-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)-3-(3-methoxy-5
(trifluoromethyl)phenyl)propanoic acid
H H O H N N N CO 2H N H N-NH 1O CF3
[0344] To a solution of 183 mg of methyl (3S)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
221102_G2572WO English Translation (Final) clean copy
indazole-6-carboxamido)acetamido)-3-(3-methoxy-5
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 5-(a) in acetonitrile (1 ml) and
water (1 ml), 24 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 2 hours.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.5 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 2 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 44 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 580 [M+H]+.
'H-NMR spectrum (400MHz, CD 3 0D) 6: 8.11 - 8.00 (m,
2H), 7.51 - 7.45 (m, 1H), 7.28 - 7.17 (m, 2H), 7.02
6.96 (m, 1H), 5.29 - 5.10 (m, 2H), 4.18 (d, J = 16.8 Hz,
1H), 4.05 (d, J = 16.8 Hz, 1H), 3.83 (s, 3H), 3.70 - 3.40
(m, 4H), 2.77 - 2.57 (m, 2H).
[0345]
Example 6-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(3-fluoro-5
(trifluoromethyl)phenyl)propanoic acid
221102_G2572WO English Translation (Final) clean copy
H H H N N N OH
F N H F N-NH F CF 3
[0346] To a solution of 163 mg of methyl (3S)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(3-fluoro-5
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 6-(a) in acetonitrile (1 ml) and
water (1 ml), 28 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 3.5 hours.
After the completion of reaction, 1 ml of water was
added to the reaction mixture. The reaction mixture was
adjusted to pH 5.6 by the addition of 1 N hydrochloric
acid, and the mixture was stirred at room temperature for
hours. A deposited solid was collected by filtration,
washed with water, and then dried under reduced pressure
at 600C to obtain 66 mg of the title compound as a white
solid.
Mass spectrum (ESI, m/z): 568 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 - 8.05 (m,
2H), 7.56 - 7.53 (m, 1H), 7.50 (d, J = 1.1 Hz, 1H), 7.46
- 7.41 (m, 1H), 7.29 - 7.24 (m, 1H), 5.30 - 5.12 (m, 2H),
4.18 - 4.05 (m, 2H), 3.70 - 3.45 (m, 4H), 2.75 - 2.60 (m,
2H).
[0347] Example 7-(b)
221102_G2572WO English Translation (Final) clean copy
Production of (3S)-3-(3,5-bis(trifluoromethyl)phenyl)-3
(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)
1H-indazole-6-carboxamido)acetamido)propanoic acid
H H H N N N OH ,UN F N-NH F3 C CF 3
[0348] To a solution of 130 mg of methyl (3S)-3-(3,5
bis(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 7-(a) in acetonitrile (1 ml) and
water (1 ml), 21 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 2.5 hours.
After the completion of reaction, 1 ml of water was
added to the reaction mixture. The reaction mixture was
adjusted to pH 5.6 by the addition of 1 N hydrochloric
acid, and the mixture was stirred at room temperature for
hours. A deposited solid was collected by filtration,
washed with water, and then dried under reduced pressure
at 600C to obtain 87 mg of the title compound as a gray
solid.
Mass spectrum (ESI, m/z): 618 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 - 8.05 (m,
2H), 8.01 - 7.98 (m, 2H), 7.81 - 7.78 (m, 1H), 7.52
7.49 (m, 1H), 5.35 (t, J= 5.3 Hz, 1H), 5.30 - 5.12 (m,
221102_G2572WO English Translation (Final) clean copy
1H), 4.16 (d, J = 16.8 Hz, 1H), 4.07 (d, J = 16.8 Hz,
1H), 3.70 - 3.45 (m, 4H), 2.78 - 2.63 (m, 2H).
[0349] Example 8-(b)
Production of (3S)-3-(3-(1H-pyrazol-1-yl)-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoic acid
H H O H N N N CO 2H
F N 0 N-NH .. F3C N N
[0350] To a solution of 150 mg of methyl (3S)-3-(3-(1H
pyrazol-1-yl)-5-(trifluoromethyl)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH
indazole-6-carboxamido)acetamido)propanoate
trifluoroacetate synthesized in Example 8-(a) in
acetonitrile (1 ml) and water (1 ml), 34 mg of lithium
hydroxide was added at room temperature under a stream of
argon, and the mixture was stirred at room temperature
for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 19 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 45 mg of
the title compound as a white solid.
221102_G2572WO English Translation (Final) clean copy
Mass spectrum (ESI, m/z): 616 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.48 (d, J = 2.4
Hz, 1H), 8.12 - 8.06 (m, 2H), 8.04 - 8.00 (m, 1H), 8.00
7.95 (m, 1H), 7.73 (d, J = 1.6 Hz, 1H), 7.66 - 7.62 (m,
1H), 7.54 (d, J = 1.0 Hz, 1H), 6.56 - 6.52 (m, 1H), 5.39
- 5.34 (m, 1H), 5.29 - 5.10 (m, 1H), 4.20 - 4.05 (m, 2H),
3.72 - 3.40 (m, 4H), 2.84 - 2.66 (m, 2H).
[0351] Example 9-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(2-fluoro-3
(trifluoromethyl)phenyl)propanoic acid
H H O H N N N CO2H F Ni NN H 0 FFO2
N-NH CF 3
[0352] To a suspension of 88 mg of methyl (3S)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(2-fluoro-3
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 9-(a) in acetonitrile (1 ml) and
water (1 ml), 16 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1.5 hours.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.4 by the addition of 1 N hydrochloric acid, and the
221102_G2572WO English Translation (Final) clean copy
mixture was stirred at room temperature for 7.5 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 60°C to
obtain 46 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 568 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.11 (d, J = 1.0
Hz, 1H), 8.08 - 8.06 (m, 1H), 7.74 - 7.69 (m, 1H), 7.56
7.51 (m, 2H), 7.27 - 7.22 (m, 1H), 5.49 (t, J = 5.2 Hz,
1H), 5.30 - 5.13 (m, 1H), 4.16 (d, J = 16.8 Hz, 1H), 4.05
(d, J = 16.8 Hz, 1H), 3.70 - 3.45 (m, 4H), 2.76 - 2.58
(m, 2H).
[0353] Example 10-(b)
Production of (3S)-3-(2-chloro-3
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
H H 0 H N N N CO 2 H H Cl F N N-NH CF 3
[0354] To a suspension of 79 mg of methyl (3S)-3-(2
chloro-3-(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 10-(a) in acetonitrile (1 ml) and
water (1 ml), 14 mg of lithium hydroxide was added at
221102_G2572WO English Translation (Final) clean copy
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1.5 hours.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.4 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 3 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 58 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 584 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.11 (d, J = 0.9
Hz, 1H), 8.10 - 8.07 (m, 1H), 7.77 - 7.73 (m, 1H), 7.66
7.62 (m, 1H), 7.56 (d, J = 1.3 Hz, 1H), 7.42 - 7.36 (m,
1H), 5.61 (t, J = 5.1 Hz, 1H), 5.29 - 5.13 (m, 1H), 4.17
- 4.03 (m, 2H), 3.70 - 3.45 (m, 4H), 2.75 - 2.58 (m, 2H).
[0355]
Example 11-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(2-fluoro-5
(trifluoromethyl)phenyl)propanoic acid
H H 0 H NN N N CO2 H 0 F F N N-NH F3 C
[0356] To a solution of 85 mg of methyl (3S)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
221102_G2572WO English Translation (Final) clean copy
indazole-6-carboxamido)acetamido)-3-(2-fluoro-5
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 11-(a) in acetonitrile (1 ml) and
water (1 ml), 15 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 3.5 hours.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.5 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 18 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 23 mg of the title compound as a pale yellow
solid.
Mass spectrum (ESI, m/z): 568 [M+H]+.
'H-NMR spectrum (400MHz, CD 3 0D) 6: 8.10 - 8.06 (m,
2H), 7.80 - 7.76 (m, 1H), 7.61 - 7.55 (m, 1H), 7.52 (d, J
= 1.0 Hz, 1H), 7.29 - 7.23 (m, 1H), 5.50 (t, J = 5.4 Hz,
1H), 5.30 - 5.14 (m, 1H), 4.16 (d, J = 16.8 Hz, 1H), 4.06
(d, J = 16.8 Hz, 1H), 3.71 - 3.45 (m, 4H), 2.78 - 2.61
(m, 2H).
[0357] Example 12-(b)
Production of (3S)-3-(2-chloro-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
221102_G2572WO English Translation (Final) clean copy
H H H N N N N CO 2H F N 0 N Cl
N-NH F 3C
[0358] To a solution of 127 mg of methyl (3S)-3-(2
chloro-5-(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 12-(a) in acetonitrile (1 ml) and
water (1 ml), 22 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.3 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 16 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 58 mg of the title compound as a gray solid.
Mass spectrum (ESI, m/z): 584 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.12 - 8.06 (m,
2H), 7.80 (d, J = 1.6 Hz, 1H), 7.59 - 7.50 (m, 3H), 5.58
(t, J = 5.3 Hz, 1H), 5.30 - 5.14 (m, 1H), 4.16 (d, J =
16.8 Hz, 1H), 4.07 (d, J= 16.8 Hz, 1H), 3.72 - 3.45 (m,
4H), 2.76 - 2.62 (m, 2H).
[0359] Example 13-(b)
221102_G2572WO English Translation (Final) clean copy
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)-3-(4-fluoro-3
(trifluoromethyl)phenyl)propanoic acid
H H 0 H N N N OH N H O
N-NH CF 3 F
[0360] To a solution of 114 mg of methyl (3S)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(4-fluoro-3
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 1-(a) in acetonitrile (1 ml) and
water (1 ml), 20 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.3 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 4 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 60°C to
obtain 62 mg of the title compound as a pale yellow
solid.
Mass spectrum (ESI, m/z): 568 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.10 - 8.05 (m,
2H), 7.72 - 7.64 (m, 2H), 7.51 (d, J = 1.1 Hz, 1H), 7.26
221102_G2572WO English Translation (Final) clean copy
- 7.20 (m, 1H), 5.31 - 5.14 (m, 2H), 4.17 - 4.02 (m, 2H),
3.73 - 3.46 (m, 4H), 2.73 - 2.60 (m, 2H).
[0361] Example 14-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)-3-(4-methyl-3
(trifluoromethyl)phenyl)propanoic acid
H H O H N N N N OH N 0 0 F'U N-NH CF 3
[0362] To a solution of 115 mg of methyl (3S)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(4-methyl-3
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 14-(a) in acetonitrile (1 ml) and
water (1 ml), 20 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.4 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 3 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 46 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 564 [M+H]+.
221102_G2572WO English Translation (Final) clean copy
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 - 8.03 (m,
2H), 7.66 - 7.62 (m, 1H), 7.51 - 7.46 (m, 2H), 7.26 (d, J
= 8.0 Hz, 1H), 5.28 - 5.12 (m, 2H), 4.16 (d, J = 16.8 Hz,
1H), 4.05 (d, J = 16.8 Hz, 1H), 3.71 - 3.43 (m, 4H), 2.72
- 2.59 (m, 2H), 2.43 - 2.38 (m, 3H).
[0363] Example 15-(b)
Production of (3S)-3-(3-chloro-4-fluoro-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
H H 0 H N N N OH
F N F H 0~~ 0 N-NH C I CF3 F
[0364] To a solution of 90 mg of methyl (3S)-3-(3-chloro
4-fluoro-5-(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 15-(a) in acetonitrile (1 ml) and
water (1 ml), 15 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, 1 ml of water was
added to the reaction mixture, and the mixture was
filtered. The filtrate was adjusted to pH 5.5 by the
addition of 1 N hydrochloric acid. 1 ml of water and 1
ml of acetonitrile were added thereto, and the mixture
221102_G2572WO English Translation (Final) clean copy
was stirred at room temperature for 10 minutes. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 60°C to
obtain 51 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 602 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.11 - 8.07 (m,
2H), 7.80 (dd, J = 2.2, 6.6 Hz, 1H), 7.66 (dd, J = 2.2,
6.0 Hz, 1H), 7.52 (d, J = 1.1 Hz, 1H), 5.30 - 5.16 (m,
2H), 4.17 - 4.02 (m, 2H), 3.73 - 3.46 (m, 4H), 2.74
2.60 (m, 2H).
[0365] Example 16-(b)
Production of (3S)-3-(3-bromo-4-methoxyphenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H H N N N N OH F N 0 0
N-NH Br
[0366] To a solution of 110 mg of methyl (3S)-3-(3-bromo
4-methoxyphenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 16-(a) in acetonitrile (1 ml) and
water (1 ml), 19 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1.5 hours.
221102_G2572WO English Translation (Final) clean copy
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.2 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 16 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 62 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 590, 592 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.06 - 8.01 (m,
2H), 7.55 (d, J = 2.1 Hz, 1H), 7.45 (d, J= 1.1 Hz, 1H),
7.30 (dd, J = 2.1, 8.7 Hz, 1H), 6.90 (d, J= 8.7 Hz, 1H),
5.29 - 5.11 (m, 2H), 4.16 (d, J = 16.8 Hz, 1H), 4.05 (d,
J = 16.8 Hz, 1H), 3.80 (s, 3H), 3.67 - 3.42 (m, 4H), 2.70
- 2.57 (m, 2H).
[0367] Example 17-(b)
Production of (3S)-3-(3,5-dichlorophenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H O H N N N N OH
1N H 0
N-NH CI CI
[0368] To a suspension of 120 mg of methyl (3S)-3-(3,5
dichlorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 17-(a) in acetonitrile (1 ml) and
221102_G2572WO English Translation (Final) clean copy
water (1 ml), 22 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 3.5 hours.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.3 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 2.5 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 76 mg of
the title compound as a pale yellow solid.
Mass spectrum (ESI, m/z): 550 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.08 - 8.05 (m,
2H), 7.51 - 7.47 (m, 1H), 7.37 - 7.34 (m, 2H), 7.25 (t, J
= 1.9 Hz, 1H), 5.30 - 5.13 (m, 2H), 4.17 - 4.05 (m, 2H),
3.71 - 3.44 (m, 4H), 2.70 - 2.57 (m, 2H).
[0369] Example 18-(b)
Production of (3S)-3-(3-bromo-5-chlorophenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H 0 H N N N OH ,UN F N H F N-NH Br Cl
[0370] To a solution of 125 mg of methyl (3S)-3-(3-bromo
-chlorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
221102_G2572WO English Translation (Final) clean copy
synthesized in Example 18-(a) in acetonitrile (1 ml) and
water (1 ml), 21 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 2.5 hours.
After the completion of reaction, 2 ml of water and
0.5 ml of acetonitrile were added to the reaction
mixture, and the mixture was filtered. The filtrate was
adjusted to pH 5.8 by the addition of 1 N hydrochloric
acid, and the mixture was stirred at room temperature for
22 hours. A deposited solid was collected by filtration,
washed with water, and then dried under reduced pressure
at 600C to obtain 85 mg of the title compound as a white
solid.
Mass spectrum (ESI, m/z): 594, 596 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.08 - 8.05 (m,
2H), 7.52 - 7.48 (m, 2H), 7.41 - 7.38 (m, 2H), 5.30
5.13 (m, 2H), 4.17 - 4.05 (m, 2H), 3.71 - 3.45 (m, 4H),
2.70 - 2.56 (m, 2H).
[0371] Example 19-(b)
Production of (3S)-3-(3,5-dibromophenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H O H N N N N CO 2H
F N 0 N-NH Br Br
221102_G2572WO English Translation (Final) clean copy
[0372] To a suspension of 48 mg of methyl (3S)-3-(3,5
dibromophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 19-(a) in acetonitrile (0.5 ml)
and water (0.5 ml), 13 mg of lithium hydroxide
monohydrate was added at room temperature under a stream
of argon, and the mixture was stirred at room temperature
for 1 hour.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.9 by the addition of 1 M
hydrochloric acid, and the mixture was stirred overnight
at room temperature. A deposited solid was collected by
filtration, washed with water, and then dried under
reduced pressure at 500C to obtain 34 mg of the title
compound as a white solid.
Mass spectrum (ESI, m/z): 640 [M+H]+.
'H-NMR spectrum (400MHz, CD 3 0D) 6: 8.10 - 8.07 (m,
2H), 7.56 - 7.53 (m, 3H), 7.52 (d, J = 1.1 Hz, 1H), 5.31
- 5.10 (m, 2H), 4.18 - 4.03 (m, 2H), 3.71 - 3.45 (m, 4H),
2.71 - 2.54 (m, 2H).
[0373] Example 20-(b)
Production of (3S)-3-(3-bromo-5-iodophenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
221102_G2572WO English Translation (Final) clean copy
H H H N N N OH FN H F N-NH Br 1
[0374] To a suspension of 140 mg of methyl (3S)-3-(3
bromo-5-iodophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 20-(a) in acetonitrile (1 ml) and
water (1 ml), 21 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 3.5 hours.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 16 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 60°C to obtain 88 mg of
the title compound as a light brown solid.
Mass spectrum (ESI, m/z): 686, 688 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.11 - 8.05 (m,
2H), 7.74 - 7.70 (m, 2H), 7.57 - 7.54 (m, 1H), 7.52 (d, J
= 1.1 Hz, 1H), 5.33 - 5.15 (m, 1H), 5.13 (t, J = 5.3 Hz,
1H), 4.17 - 4.04 (m, 2H), 3.72 - 3.45 (m, 4H), 2.69
2.55 (m, 2H).
[0375] Example 21-(b)
221102_G2572WO English Translation (Final) clean copy
Production of (3S)-3-(3-chloro-5-iodophenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H O H N N N CO 2H
F N H N-NH CI 1
[0376] To a suspension of 48 mg of methyl (3S)-3-(3
chloro-5-iodophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 21-(a) in acetonitrile (0.5 ml)
and water (0.5 ml), 13 mg of lithium hydroxide
monohydrate was added at room temperature under a stream
of argon, and the mixture was stirred at room temperature
for 1 hour.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.9 by the addition of 1 M
hydrochloric acid, and the mixture was stirred overnight
at room temperature. A deposited solid was collected by
filtration, washed with water, and then dried under
reduced pressure at 50°C to obtain 38 mg of the title
compound as a white solid.
Mass spectrum (ESI, m/z): 642 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.13 - 8.05 (m,
2H), 7.71 - 7.65 (m, 1H), 7.61 - 7.56 (m, 1H), 7.53 (d, J
= 1.1 Hz, 1H), 7.44 - 7.39 (m, 1H), 5.31 - 5.11 (m, 2H),
221102_G2572WO English Translation (Final) clean copy
4.18 - 4.03 (m, 2H), 3.73 - 3.45 (m, 4H), 2.71 - 2.54 (m,
2H).
[0377] Example 22-(b)
Production of (3S)-3-(5-chloro-2-fluorophenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH
indazole-6-carboxamido)acetamido)propanoic acid
H H 0 H N N N CO2H 0 F N N-NH C1
[0378] To a solution of 203 mg of methyl (3S)-3-(5
chloro-2-fluorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 22-(a) in acetonitrile (1 ml) and
water (1 ml), 49 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 19 hours. A deposited solid was
collected by filtration. Acetonitrile/methanol = 1/9
(v/v) (0.74 ml) was added to the obtained solid, and the
mixture was stirred at 800C. Acetonitrile/methanol = 1/9
(v/v) (0.74 ml) was further added thereto, and the
mixture was stirred at room temperature for 30 minutes.
221102_G2572WO English Translation (Final) clean copy
A deposited solid was collected by filtration, washed
with water, and then dried under reduced pressure at 600C
to obtain 46 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 534 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 - 8.04 (m,
2H), 7.51 (d, J = 1.0 Hz, 1H), 7.44 (dd, J = 2.7, 6.5 Hz,
1H), 7.22 (ddd, J = 2.7, 4.3, 8.8 Hz, 1H), 7.05 (dd, J=
8.8, 9.9 Hz, 1H), 5.45 - 5.40 (m, 1H), 5.28 - 5.10 (m,
1H), 4.19 - 4.05 (m, 2H), 3.71 - 3.41 (m, 4H), 2.75
2.57 (m, 2H).
[0379] Example 23-(b)
Production of (3S)-3-(5-bromo-2-fluorophenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H O H N N N CO2H N H F
N-NH Br
[0380] To a solution of 145 mg of methyl (3S)-3-(5-bromo
2-fluorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 23-(a) in acetonitrile (1 ml) and
water (1 ml), 48 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 30 minutes.
221102_G2572WO English Translation (Final) clean copy
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 19 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 50 mg of
the title compound as a white solid.
Mass spectrum (ESI, m/z): 578, 580 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.11 - 8.06 (m,
2H), 7.58 (dd, J= 2.6, 6.7 Hz, 1H), 7.54 (d, J = 1.1 Hz,
1H), 7.37 (ddd, J= 2.6, 4.5, 8.7 Hz, 1H), 7.00 (dd, J=
8.7, 10.1 Hz, 1H), 5.45 - 5.40 (m, 1H), 5.30 - 5.11 (m,
1H), 4.18 - 4.05 (m, 2H), 3.73 - 3.43 (m, 4H), 2.76
2.57 (m, 2H).
[0381] Example 24-(b)
Production of (3S)-3-(3-(difluoromethoxy)-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
H H 0 H N N N CO2H N H F N -N F -F
N-NH F 0 CF 3
[0382] To a solution of 118 mg of methyl (3S)-3-(3
(difluoromethoxy)-5-(trifluoromethyl)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoate
221102_G2572WO English Translation (Final) clean copy
trifluoroacetate synthesized in Example 24-(a) in
acetonitrile (1 ml) and water (1 ml), 19 mg of lithium
hydroxide was added at room temperature under a stream of
argon, and the mixture was stirred at room temperature
for 2 hours.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.5 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 15 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 60°C to
obtain 77 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 616 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.10 - 8.05 (m,
2H), 7.59 - 7.56 (m, 1H), 7.51 (d, J = 0.9 Hz, 1H), 7.46
- 7.42 (m, 1H), 7.27 - 7.24 (m, 1H), 6.97 (t, J = 73.5
Hz, 1H), 5.32 - 5.11 (m, 2H), 4.15 (d, J = 16.8 Hz, 1H),
4.06 (d, J = 16.8 Hz, 1H), 3.71 - 3.42 (m, 4H), 2.75
2.60 (m, 2H).
[0383] Example 25-(b)
Production of (3S)-3-(3-chloro-5
(difluoromethoxy)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
H H O H N N N CO 2 H
F N /O F N-NH Cl O F
221102_G2572WO English Translation (Final) clean copy
[0384] To a solution of 135 mg of methyl (3S)-3-(3
chloro-5-(difluoromethoxy)phenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 25-(a) in acetonitrile (1 ml) and
water (1 ml), 23 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.5 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 15 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 55 mg of the title compound as a pale yellow
solid.
Mass spectrum (ESI, m/z): 582 [M+H]+.
'H-NMR spectrum (400MHz, CD 3 0D) 6: 8.11 - 8.06 (m,
2H), 7.51 (d, J = 1.1 Hz, 1H), 7.30 - 7.27 (m, 1H), 7.14
- 7.10 (m, 1H), 7.06 - 7.04 (m, 1H), 6.90 (t, J = 73.5
Hz, 1H), 5.32 - 5.13 (m, 2H), 4.15 - 4.06 (m, 2H), 3.74
3.45 (m, 4H), 2.78 - 2.70 (m, 2H).
[0385] Example 26-(b)
Production of (3S)-3-(3-bromo-5-(difluoromethoxy)phenyl)
3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2
yl)amino)-1H-indazole-6-carboxamido)acetamido)propanoic
acid
221102_G2572WO English Translation (Final) clean copy
H H 0 H N N N CO 2H
F N 0 F N-INH NH Br Br 0) O F
[0386] To a solution of 177 mg of methyl (3S)-3-(3-bromo
-(difluoromethoxy)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 26-(a) in acetonitrile (1 ml) and
water (1 ml), 25 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 19 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 88 mg of
the title compound as a light brown solid.
Mass spectrum (ESI, m/z): 626, 628 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.11 - 8.05 (m,
2H), 7.52 (d, J = 1.1 Hz, 1H), 7.45 - 7.41 (m, 1H), 7.20
- 7.14 (m, 2H), 6.90 (t, J = 73.5 Hz, 1H), 5.32 - 5.15
(m, 2H), 4.16 - 4.04 (m, 2H), 3.73 - 3.44 (m, 4H), 2.76
2.61 (m, 2H).
[0387] Example 27-(b)
Production of (3S)-3-(3-(difluoromethoxy)-4
fluorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
221102_G2572WO English Translation (Final) clean copy
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoic acid
H H H N N N CO 2H Yl H F N O / F
N-NH 0 F F
[0388] To a solution of 150 mg of methyl (3S)-3-(3
(difluoromethoxy)-4-fluorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 27-(a) in acetonitrile (1 ml) and
water (1 ml), 49 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 19 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 40 mg of
the title compound as a white solid.
Mass spectrum (ESI, m/z): 566 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.11 - 8.04 (m,
2H), 7.51 (d, J = 1.1 Hz, 1H), 7.31 (dd, J = 2.1, 7.4 Hz,
1H), 7.25 (ddd, J = 2.1, 4.4, 8.6 Hz, 1H), 7.14 (dd, J =
8.6, 10.4 Hz, 1H), 6.86 (t, J= 73.8 Hz, 1H), 5.30 - 5.14
221102_G2572WO English Translation (Final) clean copy
(m, 2H), 4.14 (d, J = 16.8 Hz, 1H), 4.04 (d, J= 16.8 Hz,
1H), 3.73 - 3.42 (m, 4H), 2.72 - 2.58 (m, 2H).
[0389] Example 28-(b)
Production of (3S)-3-(5-(difluoromethoxy)-2
fluorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoic acid
H H O H N N N CO2H F N~ H FO 0 IFFO IF F N-NH F 0
[0390] To a solution of 152 mg of methyl (3S)-3-(5
(difluoromethoxy)-2-fluorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 28-(a) in acetonitrile (1 ml) and
water (1 ml), 49 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 19 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 70 mg of
the title compound as a white solid.
Mass spectrum (ESI, m/z): 566 [M+H]+.
221102_G2572WO English Translation (Final) clean copy
'H-NMR spectrum (400MHz, CD 30D) 6: 8.11 - 8.06 (m,
2H), 7.53 (d, J = 1.1 Hz, 1H), 7.22 (dd, J= 2.9, 6.1 Hz,
1H), 7.11 - 6.98 (m, 2H), 6.81 (t, J = 74.3 Hz, 1H), 5.47
- 5.42 (m, 1H), 5.30 - 5.12 (m, 1H), 4.17 - 4.03 (m, 2H),
3.71 - 3.43 (m, 4H), 2.76 - 2.61 (m, 2H).
[0391] Example 29-(b)
Production of (3S)-3-(3-(difluoromethoxy)-5-(1H-pyrazol
1-yl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
H H O H N N N CO 2H
F N F O N-NH F ON N'N
[0392] To a solution of 162 mg of methyl (3S)-3-(3
(difluoromethoxy)-5-(1H-pyrazol-1-yl)phenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoate
trifluoroacetate synthesized in Example 29-(a) in
acetonitrile (1 ml) and water (1 ml), 48 mg of lithium
hydroxide was added at room temperature under a stream of
argon, and the mixture was stirred at room temperature
for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid. 3 ml of water was added thereto, and
the mixture was stirred at room temperature for 15 hours.
221102_G2572WO English Translation (Final) clean copy
A deposited solid was collected by filtration, washed
with water, and then dried under reduced pressure at 600C
to obtain 65 mg of the title compound as a pale yellow
solid.
Mass spectrum (ESI, m/z): 614 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.38 (d, J = 2.4
Hz, 1H), 8.11 - 8.06 (m, 2H), 7.69 (d, J = 1.6 Hz, 1H),
7.62 - 7.59 (m, 1H), 7.55 (d, J = 1.0 Hz, 1H), 7.46
7.43 (m, 1H), 7.13 - 7.10 (m, 1H), 6.95 (t, J = 73.9 Hz,
1H), 6.53 - 6.49 (m, 1H), 5.30 - 5.10 (m, 2H), 4.17 (d, J
= 16.7 Hz, 1H), 4.08 (d, J = 16.7 Hz, 1H), 3.70 - 3.40
(m, 4H), 2.81 - 2.64 (m, 2H).
[0393] Example 30-(b)
Production of (3S)-3-(3,5-bis(difluoromethoxy)phenyl)-3
(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)
1H-indazole-6-carboxamido)acetamido)propanoic acid
H H O H N N N N OH
N HF 0 0 FU W F 11 F N-NH F 0 0 F
[0394] To a suspension of 51 mg of methyl (3S)-3-(3,5
bis(difluoromethoxy)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 30-(a) in acetonitrile (0.5 ml)
and water (0.5 ml), 15 mg of lithium hydroxide
monohydrate was added at room temperature under a stream
221102_G2572WO English Translation (Final) clean copy
of argon, and the mixture was stirred at room temperature
for 1 hour.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.9 by the addition of 1 M
hydrochloric acid, and the mixture was stirred overnight
at room temperature. A deposited solid was collected by
filtration, washed with water, and then dried under
reduced pressure at 500C to obtain 44 mg of the title
compound as a brown solid.
Mass spectrum (ESI, m/z): 614 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.10 - 8.03 (m,
2H), 7.54 - 7.48 (m, 1H), 7.06 - 7.00 (m, 2H), 6.90 (t, J
= 73.9 Hz, 2H), 6.77 - 6.74 (m, 1H), 5.30 - 5.12 (m, 2H),
4.19 - 3.99 (m, 2H), 3.72 - 3.43 (m, 4H), 2.71 - 2.57 (m,
2H).
[0395] Example 31-(b)
Production of (3S)-3-(3-chloro-5
(trifluoromethoxy)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoic acid
H H O H N N N CO 2 H N H
N--NH C1 OCF 3
[0396] To a suspension of 70 mg of methyl (3S)-3-(3
chloro-5-(trifluoromethoxy)phenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
221102_G2572WO English Translation (Final) clean copy
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 31-(a) in acetonitrile (0.5 ml)
and water (0.5 ml), 21 mg of lithium hydroxide
monohydrate was added at room temperature under a stream
of argon, and the mixture was stirred at room temperature
for 1 hour.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.9 by the addition of 1 M
hydrochloric acid, and the mixture was stirred overnight
at room temperature. A deposited solid was collected by
filtration, washed with water, and then dried under
reduced pressure at 50°C to obtain 52 mg of the title
compound as a white solid.
Mass spectrum (ESI, m/z): 600 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.13 - 8.05 (m,
2H), 7.52 (d, J = 1.3 Hz, 1H), 7.46 - 7.42 (m, 1H), 7.29
- 7.25 (m, 1H), 7.21 - 7.18 (m, 1H), 5.34 -5.13 (m, 2H),
4.17 - 4.05 (m, 2H), 3.78 - 3.45 (m, 4H), 2.77 - 2.65 (m,
2H).
[0397] Example 32-(b)
Production of (3S)-3-(3-bromo-5
(trifluoromethoxy)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
H H 0 H N N N OH
F NH 0 0 F N-NH Br OCF 3
221102_G2572WO English Translation (Final) clean copy
[0398] To a solution of 321 mg of methyl (3S)-3-(3-bromo
-(trifluoromethoxy)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 32-(a) in acetonitrile (2.5 ml)
and water (2.5 ml), 50 mg of lithium hydroxide was added
at room temperature under a stream of argon, and the
mixture was stirred at room temperature for 3.5 hours.
After the completion of reaction, 2.5 ml of water
was added to the reaction mixture. The reaction mixture
was adjusted to pH 5.0 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 20 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 244 mg of
the title compound as a white solid.
Mass spectrum (ESI, m/z): 644, 646 [M+H]+.
'H-NMR spectrum (400MHz, CD 3 0D) 6: 8.12 - 8.06 (m,
2H), 7.61 - 7.58 (m, 1H), 7.51 (d, J = 1.1 Hz, 1H), 7.36
- 7.33 (m, 1H), 7.33 - 7.30 (m, 1H), 5.33 - 5.16 (m, 2H),
4.16 - 4.05 (m, 2H), 3.73 - 3.49 (m, 4H), 2.76 (d, J =
6.4 Hz, 2H).
[0399] Example 33-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(2-fluoro-5
(trifluoromethoxy)phenyl)propanoic acid
221102_G2572WO English Translation (Final) clean copy
H H 0 H N N N CO2H F N N F C 2 N HH 0 IF N-NH F3CO
[0400] To a solution of 270 mg of methyl (3S)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(2-fluoro-5
(trifluoromethoxy)phenyl)propanoate trifluoroacetate
synthesized in Example 33-(a) in acetonitrile (2 ml) and
water (2 ml), 45 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.2 by the addition of 1 N
hydrochloric acid. 4 ml of water was added thereto, and
the mixture was stirred at room temperature for 16 hours.
A deposited solid was collected by filtration, washed
with water, and then dried under reduced pressure at 600C
to obtain 169 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 584 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.08 - 8.05 (m,
2H), 7.50 - 7.48 (m, 1H), 7.38 - 7.34 (m, 1H), 7.20
7.12 (m, 2H), 5.49 - 5.44 (m, 1H), 5.30 - 5.14 (m, 1H),
4.18 - 4.04 (m, 2H), 3.73 - 3.41 (m, 4H), 2.78 - 2.61 (m,
2H).
[0401] Example 34-(b)
221102_G2572WO English Translation (Final) clean copy
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)-3-(4-fluoro-3
(trifluoromethoxy)phenyl)propanoic acid
H H O H N N N N OH N OH 0
N--NH OCF3 F
[0402] To a solution of 93 mg of methyl (3S)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH
indazole-6-carboxamido)acetamido)-3-(4-fluoro-3
(trifluoromethoxy)phenyl)propanoate trifluoroacetate
synthesized in Example 34-(a) in acetonitrile (1 ml) and
water (1 ml), 16 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 2.5 hours.
After the completion of reaction, 1 ml of water was
added to the reaction mixture, and the mixture was
filtered. The filtrate was adjusted to pH 5.6 by the
addition of 1 N hydrochloric acid, and the mixture was
stirred at room temperature for 18 hours. A deposited
solid was collected by filtration, washed with water, and
then dried under reduced pressure at 600C to obtain 63 mg
of the title compound as a white solid.
Mass spectrum (ESI, m/z): 584 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.08 (d, J = 1.0
Hz, 1H), 8.07 - 8.05 (m, 1H), 7.50 (d, J = 1.1 Hz, 1H),
221102_G2572WO English Translation (Final) clean copy
7.46 - 7.37 (m, 2H), 7.22 (dd, J= 8.6, 10.1 Hz, 1H),
5.31 - 5.14 (m, 2H), 4.15 (d, J= 16.8 Hz, 1H), 4.05 (d,
J = 16.8 Hz, 1H), 3.72 - 3.46 (m, 4H), 2.71 - 2.58 (m,
2H).
[0403] Example 35-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(3
(trifluoromethyl)phenyl)propanoic acid
H H 0 H N N N N OH F N H 0 0
N-NH I CF 3
[0404] To a solution of 142 mg of methyl (3S)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(3
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 35-(a) in acetonitrile (1.5 ml)
and water (1.5 ml), 26 mg of lithium hydroxide was added
at room temperature under a stream of argon, and the
mixture was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.4 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 16 hours.
The reaction mixture was concentrated under reduced
pressure, and acetonitrile was distilled off under
221102_G2572WO English Translation (Final) clean copy
reduced pressure. The residue was stirred at room
temperature for 3 hours. A deposited solid was collected
by filtration, washed with water, and then dried under
reduced pressure at 600C to obtain 63 mg of the title
compound as a pale yellow solid.
Mass spectrum (ESI, m/z): 550 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.10 - 8.04 (m,
2H), 7.73 - 7.67 (m, 1H), 7.67 - 7.61 (m, 1H), 7.53
7.45 (m, 3H), 5.31 (t, J = 5.5 Hz, 1H), 5.29 - 5.13 (m,
1H), 4.20 - 4.04 (m, 2H), 3.72 - 3.44 (m, 4H), 2.77
2.64 (m, 2H).
[0405] Example 36-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)-3-(3
(trifluoromethoxy)phenyl)propanoic acid
H H O H N N NyN -- OH
N OH 0
N-NH OCF 3
[0406] To a solution of 104 mg of methyl (3S)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(3
(trifluoromethoxy)phenyl)propanoate trifluoroacetate
synthesized in Example 36-(a) in acetonitrile (1 ml) and
water (1 ml), 18 mg of lithium hydroxide was added at
221102_G2572WO English Translation (Final) clean copy
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.2 by the addition of 1 N hydrochloric acid. The
reaction mixture was concentrated under reduced pressure,
and acetonitrile was distilled off under reduced
pressure. The residue was stirred at room temperature
for 2 hours. A deposited solid was collected by
filtration, washed with water, and then dried under
reduced pressure at 60°C to obtain 50 mg of the title
compound as a pale yellow solid.
Mass spectrum (ESI, m/z): 566 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.08 (d, J = 0.9
Hz, 1H), 8.06 - 8.04 (m, 1H), 7.48 (d, J = 1.1 Hz, 1H),
7.41 - 7.36 (m, 2H), 7.32 - 7.28 (m, 1H), 7.15 - 7.09 (m,
1H), 5.33 - 5.14 (m, 2H), 4.18 - 4.04 (m, 2H), 3.71
3.45 (m, 4H), 2.78 - 2.66 (m, 2H).
[0407] Example 37-(b)
Production of (3S)-3-(3-(difluoromethoxy)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H O H N N N N OH
F N O OF N-NH 0 F
221102_G2572WO English Translation (Final) clean copy
[0408] To a solution of 110 mg of methyl (3S)-3-(3
(difluoromethoxy)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 37-(a) in acetonitrile (1 ml) and
water (1 ml), 20 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.5 by the addition of 1 N hydrochloric acid. The
reaction mixture was concentrated under reduced pressure,
and acetonitrile was distilled off under reduced
pressure. The residue was stirred at room temperature
for 2.5 hours. A deposited solid was collected by
filtration, washed with water, and then dried under
reduced pressure at 600C to obtain 50 mg of the title
compound as a beige solid.
Mass spectrum (ESI, m/z): 548 [M+H]+.
'H-NMR spectrum (400MHz, CD 3 0D) 6: 8.10 - 8.03 (m,
2H), 7.50 (d, J = 1.1 Hz, 1H), 7.32 - 7.27 (m, 1H), 7.26
- 7.21 (m, 1H), 7.18 - 7.15 (m, 1H), 6.98 - 6.93 (m, 1H),
6.83 (t, J = 74.3 Hz, 1H), 5.29 - 5.12 (m, 2H), 4.19
4.02 (m, 2H), 3.70 - 3.43 (m, 4H), 2.72 - 2.60 (m, 2H).
[0409] Example 38-(b)
221102_G2572WO English Translation (Final) clean copy
Production of (3S)-3-(3-(difluoromethyl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H 0 H N N N N OH
F N O O N-NH F F
[0410] To a solution of 150 mg of methyl (3S)-3-(3
(difluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 38-(a) in acetonitrile (1 ml) and
water (1 ml), 28 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 2.5 hours.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.3 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 18 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 60°C to
obtain 60 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 532 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.06 (d, J = 1.0
Hz, 1H), 8.05 - 8.02 (m, 1H), 7.57 - 7.51 (m, 2H), 7.47
(d, J = 1.3 Hz, 1H), 7.42 - 7.35 (m, 2H), 6.71 (t, J =
56.2 Hz, 1H), 5.30 - 5.11 (m, 2H), 4.17 (d, J = 16.8 Hz,
221102_G2572WO English Translation (Final) clean copy
1H), 4.06 (d, J = 16.8 Hz, 1H), 3.68 - 3.41 (m, 4H), 2.75
- 2.63 (m, 2H).
[0411] Example 39-(b)
Production of (3S)-3-(3-cyclopropylphenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH
indazole-6-carboxamido)acetamido)propanoic acid
H H 0 H N N N CO 2H N H
N-NH
[0412] To a solution of 149 mg of methyl (3S)-3-(3
cyclopropylphenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 39-(a) in acetonitrile (1 ml) and
water (1 ml), 25 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 19 hours. A deposited solid was
collected by filtration, washed with water, and then
dried under reduced pressure at 600C to obtain 66 mg of
the title compound as a white solid.
Mass spectrum (ESI, m/z): 522 [M+H]+.
221102_G2572WO English Translation (Final) clean copy
'H-NMR spectrum (400MHz, CD 30D) 6: 8.06 (d, J = 1.0
Hz, 1H), 8.03 (dd, J = 1.0, 1.1 Hz, 1H), 7.47 (d, J = 1.1
Hz, 1H), 7.14 - 7.07 (m, 3H), 6.89 - 6.85 (m, 1H), 5.27
5.11 (m, 2H), 4.19 (d, J = 16.7 Hz, 1H), 4.03 (d, J =
16.7 Hz, 1H), 3.70 - 3.38 (m, 4H), 2.71 - 2.58 (m, 2H),
1.88 - 1.79 (m, 1H), 0.93 - 0.83 (m, 2H), 0.70 - 0.59 (m,
2H).
[0413] Example 40-(b)
Production of (3S)-3-(3-cyclopropoxyphenyl)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H H N N N CO2H
F N N-NH O
[0414] To a solution of 155 mg of methyl (3S)-3-(3
cyclopropoxyphenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 40-(a) in acetonitrile (1 ml) and
water (1 ml), 25 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 15 hours. A deposited solid was
collected by filtration, washed with water, and then
221102_G2572WO English Translation (Final) clean copy
dried under reduced pressure at 600C to obtain 51 mg of
the title compound as a white solid.
Mass spectrum (ESI, m/z): 538 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.06 (d, J = 0.9
Hz, 1H), 8.02 (dd, J = 0.9, 1.1 Hz, 1H), 7.45 (d, J= 1.1
Hz, 1H), 7.16 (dd, J = 7.7, 7.8 Hz, 1H), 7.06 (dd, J=
1.9, 2.1 Hz, 1H), 6.98 - 6.94 (m, 1H), 6.87 (dd, J = 2.1,
7.8 Hz, 1H), 5.27 - 5.11 (m, 2H), 4.19 (d, J = 16.8 Hz,
1H), 4.04 (d, J = 16.8 Hz, 1H), 3.73 (tt, J = 3.0, 6.0
Hz, 1H), 3.68 - 3.40 (m, 4H), 2.72 - 2.60 (m, 2H), 0.79
0.55 (m, 4H).
[0415] Example 41-(b)
Production of (3S)-3-(3-chlorophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
H H O H N N N N OH F IN O O
N-NH C1
[0416] To a solution of 99 mg of methyl (3S)-3-(3
chlorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 41-(a) in acetonitrile (1 ml) and
water (1 ml), 19 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 4 hours.
221102_G2572WO English Translation (Final) clean copy
After the completion of reaction, 1 ml of water was
added to the reaction mixture, and the mixture was
filtered. The filtrate was adjusted to pH 5.4 by the
addition of 1 N hydrochloric acid, and the mixture was
stirred at room temperature for 4 hours. A deposited
solid was collected by filtration, washed with water, and
then dried under reduced pressure at 600C to obtain 33 mg
of the title compound as a white solid.
Mass spectrum (ESI, m/z): 516 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.08 (d, J = 1.0
Hz, 1H), 8.07 - 8.06 (m, 1H), 7.50 (d, J = 1.3 Hz, 1H),
7.42 - 7.39 (m, 1H), 7.32 - 7.28 (m, 1H), 7.25 (t, J =
7.7 Hz, 1H), 7.21 - 7.17 (m, 1H), 5.29 - 5.13 (m, 2H),
4.18 - 4.04 (m, 2H), 3.71 - 3.44 (m, 4H), 2.71 - 2.59 (m,
2H).
[0417] Example 42-(b)
Production of (3S)-3-(3-bromophenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid
H H 0 H N N N OH F H 0 0 F N--NH Br
[0418] To a suspension of 122 mg of methyl (3S)-3-(3
bromophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
221102_G2572WO English Translation (Final) clean copy
synthesized in Example 42-(a) in acetonitrile (1 ml) and
water (1 ml), 22 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 2 hours.
After the completion of reaction, 1 ml of water was
added to the reaction mixture, and the mixture was
filtered. The filtrate was adjusted to pH 5.3 by the
addition of 1 N hydrochloric acid, and the mixture was
stirred at room temperature for 2 hours. A deposited
solid was collected by filtration, washed with water, and
then dried under reduced pressure at 600C to obtain 66 mg
of the title compound as a white solid.
Mass spectrum (ESI, m/z): 560, 562 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 - 8.06 (m,
2H), 7.56 (t, J = 1.7 Hz, 1H), 7.52 (d, J = 1.1 Hz, 1H),
7.36 - 7.32 (m, 2H), 7.22 - 7.17 (m, 1H), 5.30 - 5.14 (m,
2H), 4.18 - 4.04 (m, 2H), 3.71 - 3.45 (m, 4H), 2.70
2.57 (m, 2H).
[0419] Example 43-(b)
Production of (3S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H H N N N N CO 2H
F F N O N--NH -N N2
221102_G2572WO English Translation (Final) clean copy
[0420] To a solution of 123 mg of methyl (3S)-3-(3-(1H
pyrazol-1-yl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 43-(a) in acetonitrile (1 ml) and
water (1 ml), 25 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 19 hours. The reaction mixture was
purified by BondElut (eluting solvent:
water:acetonitrile:methanol) to obtain 49 mg of the title
compound as a white solid.
Mass spectrum (ESI, m/z): 548 [M+H]+.
'H-NMR spectrum (400MHz, CD 3 0D) 6: 8.30 (dd, J = 0.5,
2.5 Hz, 1H), 8.08 (d, J = 0.8 Hz, 1H), 8.06 (dd, J = 0.8,
1.3 Hz, 1H), 7.74 - 7.70 (m, 1H), 7.68 - 7.67 (m, 1H),
7.61 - 7.55 (m, 1H), 7.52 (d, J = 1.3 Hz, 1H), 7.44
7.31 (m, 2H), 6.49 (dd, J = 1.9, 2.5 Hz, 1H), 5.33 - 5.28
(m, 1H), 5.25 - 5.07 (m, 1H), 4.21 - 4.06 (m, 2H), 3.66
3.37 (m, 4H), 2.84 - 2.65 (m, 2H).
[0421] Example 44-(b)
Production of (3S)-3-(3-(3,5-dimethyl-1H-pyrazol-1
yl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
221102_G2572WO English Translation (Final) clean copy
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoic acid
H H 0 H N N N CO 2 H
F N H N-NH N
[0422] To a solution of 120 mg of methyl (3S)-3-(3-(3,5
dimethyl-1H-pyrazol-1-yl)phenyl)-3-(2-(4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 44-(a) in acetonitrile (1 ml) and
water (1 ml), 41 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.0 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 1 hour.
The reaction mixture was concentrated under reduced
pressure, and acetonitrile was distilled off under
reduced pressure. 300 ptl of acetonitrile was added to
the residue, and the mixture was stirred at room
temperature for 1 hour. A deposited solid was collected
by filtration, washed with water, and then dried under
reduced pressure at 600C to obtain 35 mg of the title
compound as a white solid.
Mass spectrum (ESI, m/z): 576 [M+H]+.
221102_G2572WO English Translation (Final) clean copy
'H-NMR spectrum (400MHz, CD 30D) 6: 8.03 (d, J = 0.9
Hz, 1H), 8.00 - 7.98 (m, 1H), 7.47 - 7.39 (m, 4H), 7.29
7.22 (m, 1H), 6.00 (s, 1H), 5.33 - 5.28 (m, 1H), 5.26
5.08 (m, 1H), 4.17 (d, J = 16.8 Hz, 1H), 4.06 (d, J =
16.8 Hz, 1H), 3.66 - 3.38 (m, 4H), 2.82 - 2.60 (m, 2H),
2.23 (s, 3H), 2.19 (s, 3H).
[0423] Example 45-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(3-morpholinophenyl)propanoic
acid
H H 0 H N N N CO2H
F N O N--NH N O
[0424] To a solution of 78 mg of methyl (3S)-3-(2-(4-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(3
morpholinophenyl)propanoate trifluoroacetate synthesized
in Example 45-(a) in acetonitrile (1 ml) and water (1
ml), 27 mg of lithium hydroxide was added at room
temperature under a stream of argon, and the mixture was
stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.5 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 19 hours. The reaction mixture was
221102_G2572WO English Translation (Final) clean copy
purified by BondElut (eluting solvent:
water:acetonitrile:methanol) to obtain 44 mg of the title
compound as a white solid.
Mass spectrum (ESI, m/z): 567 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.08 (d, J = 0.9
Hz, 1H), 8.03 (dd, J = 0.9, 1.1 Hz, 1H), 7.48 (d, J = 1.1
Hz, 1H), 7.18 - 7.12 (m, 1H), 6.99 - 6.96 (m, 1H), 6.89
6.85 (m, 1H), 6.80 - 6.76 (m, 1H), 5.28 - 5.11 (m, 2H),
4.20 (d, J = 16.7 Hz, 1H), 4.01 (d, J = 16.7 Hz, 1H),
3.80 - 3.74 (m, 4H), 3.69 - 3.39 (m, 4H), 3.14 - 3.06 (m,
4H), 2.79 - 2.57 (m, 2H).
[0425] Example 46-(b)
Production of (3S)-3-(4-(difluoromethoxy)phenyl)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
H H 0 H
F N N 0 OH N-NH
O F F
[0426] To a solution of 126 mg of methyl (3S)-3-(4
(difluoromethoxy)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 46-(a) in acetonitrile (1 ml) and
water (1 ml), 23 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
221102_G2572WO English Translation (Final) clean copy
After the completion of reaction, 1 ml of water was
added to the reaction mixture, and the mixture was
filtered. The filtrate was adjusted to pH 5.5 by the
addition of 1 N hydrochloric acid, and the mixture was
stirred at room temperature for 17 hours. A deposited
solid was collected by filtration, washed with water, and
then dried under reduced pressure at 600C to obtain 69 mg
of the title compound as a white solid.
Mass spectrum (ESI, m/z): 548 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 (d, J= 1.0
Hz, 1H), 8.06 - 8.04 (m, 1H), 7.50 (d, J = 1.3 Hz, 1H),
7.42 - 7.38 (m, 2H), 7.06 - 7.02 (m, 2H), 6.73 (t, J =
74.3 Hz, 1H), 5.28 - 5.13 (m, 2H), 4.19 - 4.01 (m, 2H),
3.70 - 3.42 (m, 4H), 2.71 - 2.59 (m, 2H).
[0427] Example 47-(b)
Production of (3S)-3-(2-(4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)-3-(4
(trifluoromethyl)phenyl)propanoic acid
H H 0 H N N N N OH
N N-NH CF 3
[0428] To a solution of 125 mg of methyl (3S)-3-(2-(4
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)-3-(4
(trifluoromethyl)phenyl)propanoate trifluoroacetate
221102_G2572WO English Translation (Final) clean copy
synthesized in Example 47-(a) in acetonitrile (1 ml) and
water (1 ml), 23 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 2.5 hours.
After the completion of reaction, 1 ml of water was
added to the reaction mixture, and the mixture was
filtered. The filtrate was adjusted to pH 5.4 by the
addition of 1 N hydrochloric acid, and the mixture was
stirred at room temperature for 4 hours. A deposited
solid was collected by filtration, washed with water, and
then dried under reduced pressure at 600C to obtain 70 mg
of the title compound as a white solid.
Mass spectrum (ESI, m/z): 550 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.09 (d, J = 1.0
Hz, 1H), 8.07 - 8.05 (m, 1H), 7.56 (s, 4H), 7.52 (d, J=
1.1 Hz, 1H), 5.30 - 5.13 (m, 2H), 4.17 (d, J = 16.8 Hz,
1H), 4.07 (d, J = 16.8 Hz, 1H), 3.71 - 3.43 (m, 4H), 2.74
- 2.62 (m, 2H).
[0429] Example 48-(b)
Production of (3S)-3-(3-chloro-5
(trifluoromethyl)phenyl)-3-(2-(3-fluoro-4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoic acid F
H
FH i FC,
221102_G2572WO English Translation (Final) clean copy
[0430] To a solution of 65 mg of methyl (3S)-3-(3-chloro
-(trifluoromethyl)phenyl)-3-(2-(3-fluoro-4-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 48-(a) in acetonitrile (0.8 ml)
and water (0.8 ml), 10 mg of lithium hydroxide was added
at room temperature under a stream of argon, and the
mixture was stirred at room temperature for 30 minutes.
Subsequently, 10 mg of lithium hydroxide was further
added thereto at room temperature, and the mixture was
stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.6 by the addition of 1 N
hydrochloric acid. 1.6 ml of water was added thereto,
and the mixture was stirred at room temperature for 16
hours. A deposited solid was collected by filtration,
washed with water, and then dried under reduced pressure
at 600C to obtain 38 mg of the title compound as a white
solid.
Mass spectrum (ESI, m/z): 602 [M+H]+.
'H-NMR spectrum (400MHz, CD 3 0D) 6: 8.01 - 7.94 (m,
1H), 7.70 - 7.61 (m, 2H), 7.55 - 7.49 (m, 1H), 7.46
7.41 (m, 1H), 5.31 - 5.08 (m, 2H), 4.18 - 4.03 (m, 2H),
3.70 - 3.41 (m, 4H), 2.79 - 2.56 (m, 2H).
[0431] Example 49-(b)
Production of (3S)-3-(2-(3-chloro-4-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
221102_G2572WO English Translation (Final) clean copy
carboxamido)acetamido)-3-(3-chloro-5
(trifluoromethyl)phenyl)propanoic acid F
HN N o OH C1HN I~ N( CIH OH N-NH CI CF3
[0432] To a solution of 54 mg of methyl (3S)-3-(2-(3
chloro-4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2
yl)amino)-lH-indazole-6-carboxamido)acetamido)-3-(3
chloro-5-(trifluoromethyl)phenyl)propanoate
trifluoroacetate synthesized in Example 49-(a) in
acetonitrile (0.5 ml) and water (0.5 ml), 10 mg of
lithium hydroxide was added at room temperature under a
stream of argon, and the mixture was stirred at room
temperature for 1 hour.
After the completion of reaction, 1 ml of water was
added to the reaction mixture. The reaction mixture was
adjusted to pH 5.6 by the addition of 1 N hydrochloric
acid, and the mixture was stirred at room temperature for
22 hours. A deposited solid was collected by filtration,
washed with water, and then dried under reduced pressure
at 600C to obtain 36 mg of the title compound as a white
solid.
Mass spectrum (ESI, m/z): 618 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.14 (d, J = 1.3
Hz, 1H), 7.69 - 7.67 (m, 1H), 7.65 - 7.62 (m, 1H), 7.56
(d, J = 1.1 Hz, 1H), 7.54 - 7.51 (m, 1H), 5.27 - 5.10 (m,
221102_G2572WO English Translation (Final) clean copy
2H), 4.17 - 4.04 (m, 2H), 3.68 - 3.44 (m, 4H), 2.73
2.59 (m, 2H).
[0433] Example 50-(b)
Production of (3S)-3-(3-chloro-5
(trifluoromethyl)phenyl)-3-(2-(4-((5-hydroxy-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-6
carboxamido)acetamido)propanoic acid
H H 0 H N NN OH
NHH HOC1 CF 3 N-NH C ~
[0434] To a solution of 115 mg of methyl (3S)-3-(3
chloro-5-(trifluoromethyl)phenyl)-3-(2-(4-((5-hydroxy
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 50-(a) in acetonitrile (1 ml) and
water (1 ml), 20 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1.5 hours.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.6 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 4 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 72 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 582 [M+H]+.
221102_G2572WO English Translation (Final) clean copy
'H-NMR spectrum (400MHz, CD 30D) 6: 8.16 (d, J = 1.0
Hz, 1H), 8.06 - 8.04 (m, 1H), 7.70 - 7.67 (m, 1H), 7.66
7.63 (m, 1H), 7.53 - 7.51 (m, 1H), 7.50 (d, J = 1.1 Hz,
1H), 5.26 (t, J = 5.3 Hz, 1H), 4.24 (quin, J = 3.1 Hz,
1H), 4.18 - 4.05 (m, 2H), 3.49 - 3.38 (m, 2H), 3.36
3.29 (m, 2H), 2.74 - 2.60 (m, 2H).
[0435] Example 51-(b)
Production of (S)-3-(3-chloro-5-(trifluoromethyl)phenyl)
3-(2-(4-((1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-6-carboxamido)acetamido)propanoic acid
NON CH O H -N0 0 N-NH C1 Fa
[0436] To a suspension of 117 mg of methyl (S)-3-(3
chloro-5-(trifluoromethyl)phenyl)-3-(2-(4-((1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 51-(a) in acetonitrile (1 ml) and
water (1 ml), 21 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.6 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 2 hours. A
deposited solid was collected by filtration, washed with
221102_G2572WO English Translation (Final) clean copy
water, and then dried under reduced pressure at 600C to
obtain 47 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 566 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.12 (d, J = 1.0
Hz, 1H), 8.04 - 8.02 (m, 1H), 7.71 - 7.68 (m, 1H), 7.67
7.64 (m, 1H), 7.54 - 7.51 (m, 1H), 7.49 (d, J = 1.1 Hz,
1H), 5.26 (t, J = 5.2 Hz, 1H), 4.16 (d, J = 16.8 Hz, 1H),
4.06 (d, J = 16.8 Hz, 1H), 3.40 - 3.26 (m, 4H), 2.75
2.59 (m, 2H), 2.00 - 1.90 (m, 2H).
[0437] Example 52-(b)
Production of (3S)-3-(3-chloro-5
(trifluoromethyl)phenyl)-3-(2-(6-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-4
carboxamido)acetamido)propanoic acid
H H 0 H N N N OH
F NN H0 0 F / O y HN-N CI CF3
[0438] To a suspension of 152 mg of methyl (3S)-3-(3
chloro-5-(trifluoromethyl)phenyl)-3-(2-(6-((5-fluoro
1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-4
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 52-(a) in acetonitrile (2 ml) and
water (2 ml), 26 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 2 hours.
221102_G2572WO English Translation (Final) clean copy
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.8 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 4 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 86 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 584 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.48 - 8.45 (m,
1H), 7.71 - 7.68 (m, 1H), 7.66 - 7.64 (m, 1H), 7.62 (d, J
= 1.6 Hz, 1H), 7.61 - 7.58 (m, 1H), 7.54 - 7.51 (m, 1H),
5.29 - 5.12 (m, 2H), 4.19 - 4.10 (m, 2H), 3.72 - 3.46 (m,
4H), 2.72 - 2.59 (m, 2H).
[0439] Example 53-(b)
Production of (3S)-3-(3-bromo-5-(trifluoromethyl)phenyl)
3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2
yl)amino)-1H-indazole-4-carboxamido)acetamido)propanoic
acid
H H 0 H N N N CO 2 H
F N 0 HN-N Br CF 3
[0440] To a solution of 192 mg of methyl (3S)-3-(3-bromo
-(trifluoromethyl)phenyl)-3-(2-(6-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-4
carboxamido)acetamido)propanoate trifluoroacetate
synthesized in Example 53-(a) in acetonitrile (3 ml) and
221102_G2572WO English Translation (Final) clean copy
water (3 ml), 30 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 30 minutes.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.8 by the addition of 1 N
hydrochloric acid, and the mixture was stirred at room
temperature for 2 hours. A deposited solid was collected
by filtration, washed with water, and then dried under
reduced pressure at 600C to obtain 84 mg of the title
compound as a white solid.
Mass spectrum (ESI, m/z): 628, 630 [M+H]+.
'H-NMR spectrum (400MHz, DMSO-d 6 +D 20) 6: 8.35 - 8.31
(m, 1H), 7.86 - 7.83 (m, 1H), 7.79 - 7.67 (m, 2H), 7.54
7.49 (m, 1H), 7.44 - 7.40 (m, 1H), 5.32 - 5.06 (m, 2H),
4.07 - 3.89 (m, 2H), 3.60 - 3.29 (m, 4H), 2.62 - 2.40 (m,
2H).
[0441] Example 54-(b)
Production of (3S)-3-(2-(6-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-4
carboxamido)acetamido)-3-(3-iodo-5
(trifluoromethyl)phenyl)propanoic acid
H H 0 H N N N OH
H O F N HN-N CF3
[0442] To a solution of 85 mg of methyl (3S)-3-(2-(6-((5
fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
221102_G2572WO English Translation (Final) clean copy
indazole-4-carboxamido)acetamido)-3-(3-iodo-5
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 54-(a) in acetonitrile (1 ml) and
water (1 ml), 13 mg of lithium hydroxide was added at
room temperature under a stream of argon, and the mixture
was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was adjusted to pH 5.4 by the addition of 1 N
hydrochloric acid, and the mixture was filtered. The
filtrate was concentrated under reduced pressure, and
acetonitrile was distilled off under reduced pressure.
The residue was stirred at room temperature for 5 hours.
A deposited solid was collected by filtration, washed
with water, and then dried under reduced pressure at 600C
to obtain 38 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 676 [M+H]+.
'H-NMR spectrum (400MHz, CD 3 0D) 6: 8.48 - 8.47 (m,
1H), 8.03 - 8.01 (m, 1H), 7.84 - 7.82 (m, 1H), 7.72
7.69 (m, 1H), 7.62 (d, J = 1.6 Hz, 1H), 7.60 - 7.58 (m,
1H), 5.27 - 5.12 (m, 2H), 4.20 - 4.08 (m, 2H), 3.72
3.46 (m, 4H), 2.71 - 2.57 (m, 2H).
[0443] Example 55-(b)
Production of (3S)-3-(2-(6-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-1H-indazole-4
carboxamido)acetamido)-3-(4-fluoro-3
(trifluoromethyl)phenyl)propanoic acid
221102_G2572WO English Translation (Final) clean copy
H H 0 H N NN n N OOH
F N H0 0 HN-N CF 3 F
[0444] To a suspension of 142 mg of methyl (3S)-3-(2-(6
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-4-carboxamido)acetamido)-3-(4-fluoro-3
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 55-(a) in acetonitrile (1.5 ml)
and water (1.5 ml), 25 mg of lithium hydroxide was added
at room temperature under a stream of argon, and the
mixture was stirred at room temperature for 1.5 hours.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.5 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 2 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 77 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 568 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.45 - 8.43 (m,
1H), 7.73 - 7.64 (m, 2H), 7.62 - 7.59 (m, 2H), 7.27
7.20 (m, 1H), 5.28 - 5.13 (m, 2H), 4.20 - 4.08 (m, 2H),
3.72 - 3.46 (m, 4H), 2.71 - 2.57 (m, 2H).
[0445] Example 56-(b)
Production of (3S)-3-(2-(6-((5-fluoro-1,4,5,6
tetrahydropyrimidin-2-yl)amino)-lH-indazole-4
221102_G2572WO English Translation (Final) clean copy
carboxamido)acetamido)-3-(3
(trifluoromethyl)phenyl)propanoic acid
H H 0 H N N N N OH
O O F N HN-N CF 3
[0446] To a suspension of 161 mg of methyl (3S)-3-(2-(6
((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
indazole-4-carboxamido)acetamido)-3-(3
(trifluoromethyl)phenyl)propanoate trifluoroacetate
synthesized in Example 56-(a) in acetonitrile (1.5 ml)
and water (1.5 ml), 28 mg of lithium hydroxide was added
at room temperature under a stream of argon, and the
mixture was stirred at room temperature for 1 hour.
After the completion of reaction, the reaction
mixture was filtered. The filtrate was adjusted to pH
5.8 by the addition of 1 N hydrochloric acid, and the
mixture was stirred at room temperature for 3 hours. A
deposited solid was collected by filtration, washed with
water, and then dried under reduced pressure at 600C to
obtain 96 mg of the title compound as a white solid.
Mass spectrum (ESI, m/z): 550 [M+H]+.
'H-NMR spectrum (400MHz, CD 30D) 6: 8.45 - 8.43 (m,
1H), 7.72 - 7.68 (m, 1H), 7.67 - 7.63 (m, 1H), 7.62 (d, J
= 1.6 Hz, 1H), 7.60 - 7.58 (m, 1H), 7.50 - 7.46 (m, 2H),
5.32 - 5.27 (m, 1H), 5.27 - 5.11 (m, 1H), 4.21 - 4.09 (m,
2H), 3.71 - 3.46 (m, 4H), 2.73 - 2.60 (m, 2H).
221102_G2572W0 English Translation (Final) clean copy
[0447] (Test Example 1) axvfl cell adhesion inhibition
test
Test Example 1 was performed by partially modifying
the method of Reed et al. (Sci Transl Med., 7 (288),
288ra79, 2015).
Human fibronectin (manufactured by Sigma-Aldrich Co.
LLC, F0895) was prepared at 1.25 pg/mL with phosphate
buffered saline (PBS), dispensed at 100 pL/well to a 96
well plate, and left standing overnight. The plate was
washed with Hanks' balanced salt solution (HBSS), then
subjected to blocking treatment with Dulbecco's modified
Eagle medium (DMEM) containing 1% bovine serum-derived
albumin (BSA) (FUJIFILM Wako Pure Chemical Corp., for
cell culture, 017-22231), and used as a coated plate in
the adhesion test.
[0448] axv-transfected cells (CHO cells deficient in u5
gene and transfected with av gene) cultured in Ham's F-12
medium (FUJIFILM Wako Pure Chemical Corp., 087-08335)
containing 10% fetal bovine serum (FBS), 1%
penicillin/streptomycin/amphotericin B (manufactured by
Thermo Fisher Scientific, Inc., 15240), 1 mM GlutaMAX
Supplement (manufactured by Thermo Fisher Scientific,
Inc., 35050), and 400 pg/mL hygromycin B (FUJIFILM Wako
Pure Chemical Corp., 080-07683) were recovered and
suspended in DMEM containing 1 mM MnCl 2 (manufactured by
Sigma-Aldrich Co. LLC, M1787) to prepare a cell
221102_G2572WO English Translation (Final) clean copy
suspension. A test compound dissolved in DMSO was added
thereto.
[0449] The cell suspension of 1 x 106 cells/mL containing
the test compound was added at 100 pL/well to the coated
plate and left standing for 1 hour in a 5% C02 incubator
(37°C) . One hour later, the supernatant was removed, and
the plate was washed with HBSS. The cells that adhered
to each well in the plate were stained with crystal
violet, and the cells were lysed, followed by absorbance
measurement. Inhibition without substrate coating was
defined as 100% inhibition, and inhibition when DMSO was
added instead of the test compound was defined as 0%
inhibition. The rate of inhibition of cell adhesion by
the test compound was calculated according to the
following expression.
Rate of inhibition of cell adhesion (%) = [1
{(Absorbance in the case of adding the test compound)
(Absorbance without substrate coating)} / {(Absorbance in
the case of adding DMSO) - (Absorbance without substrate
coating)}] x 100
[0450] An IC50 value was calculated from the rate of
inhibition of cell adhesion by the test compound. The
results about each test compound in this test are shown
in Table 18 below. A: IC50 < 3 nM, B: 3 nM < IC50 < 10 nM,
C: 10 nM < IC50 < 30 nM, D: 30 nM < IC50 < 100 nM, or E:
IC5o > 100 nM.
221102_G2572WO English Translation (Final) clean copy
Table 18. Results of avp1 cell adhesion inhibition test about compound of Example Example No. IC 50 Example No. IC 50 Example No. IC50 1-(b) A 21-(b) A 41-(b) B 2-(b) B 22-(b) C 42-(b) B 3-(b) B 23-(b) A 43-(b) B 4-(b) B 24-(b) A 44-(b) A -(b) A 25-(b) A 45-(b) A 6-(b) A 26-(b) A 46-(b) B 7-(b) A 27-(b) A 47-(b) B 8-(b) A 28-(b) A 48-(b) B 9-(b) B 29-(b) A 49-(b) A 10-(b) E 30-(b) A 50-(b) C 11-(b) B 31-(b) B 51-(b) D 12-(b) B 32-(b) A 52-(b) A 13-(b) A 33-(b) A 53-(b) A 14-(b) A 34-(b) A 54-(b) A 15-(b) A 35-(b) B 55-(b) A 16-(b) A 36-(b) B 56-(b) B 17-(b) A 37-(b) B 18-(b) A 38-(b) A 19-(b) A 39-(b) A 20-(b) A 40-(b) A
[0451] (Test Example 2) axvf6 cell adhesion inhibition
test
Test Example 2 was performed by partially modifying
the method of Henderson et al. (Nat Med., 19 (12), 1617
24, 2013). In Test Example 2, the same reagents as in
Test Example 1 were used unless otherwise specified.
Recombinant human LAP protein (manufactured by R&D
Systems, Inc., 246-LP-025) was prepared at 0.2 pg/mL with
221102_G2572WO English Translation (Final) clean copy
PBS, dispensed at 100 pL/well to a 96-well plate, and
left standing overnight. The plate was washed with HBSS,
then subjected to blocking treatment with DMEM containing
1% BSA, and used as a coated plate in the adhesion test.
[0452] HT-29 cells cultured in McCoy's 5A medium
(manufactured by Thermo Fisher Scientific, Inc., 16600)
containing 10% FBS and 1%
penicillin/streptomycin/amphotericin B were recovered and
suspended in DMEM containing 0.1% BSA and 200 pM MnCl 2 to
prepare a cell suspension. A test compound was added
thereto.
[0453] The cell suspension of 5 x 105 cells/mL containing
the test compound was added at 100 pL/well to the coated
plate and left standing for 1 hour in a 5% C02 incubator
(37°C) . One hour later, the supernatant was removed, and
the plate was washed with HBSS containing 200 pM MnCl 2 . The cells that adhered to each well in the plate were
stained with crystal violet, and the cells were lysed,
followed by absorbance measurement. Inhibition without
substrate coating was defined as 100% inhibition, and
inhibition when DMSO was added instead of the test
compound was defined as 0% inhibition. The rate of
inhibition of cell adhesion by the test compound was
calculated according to the following expression.
Rate of inhibition of cell adhesion (%) = [1
{(Absorbance in the case of adding the test compound)
(Absorbance without substrate coating)} / {(Absorbance in
221102_G2572WO English Translation (Final) clean copy
the case of adding DMSO) - (Absorbance without substrate
coating)}] x 100
[0454] An IC50 value was calculated from the rate of
inhibition of cell adhesion by the test compound. The
results about each test compound in this test are shown
in Table 19 below. A: IC50 < 3 nM, B: 3 nM < IC50 < 10 nM,
C: 10 nM < IC50 < 30 nM, D: 30 nM < IC50 < 100 nM, or E:
IC50 100 nM.
Table 19. Results of avp6 cell adhesion inhibition test about compound of Example Example No. IC50 Example No. IC50 Example No. IC 50 1-(b) A 21-(b) A 41-(b) A 2-(b) A 22-(b) B 42-(b) A 3-(b) A 23-(b) B 43-(b) A 4-(b) A 24-(b) A 44-(b) A -(b) A 25-(b) A 45-(b) B 6-(b) B 26-(b) A 46-(b) B 7-(b) A 27-(b) B 47-(b) B 8-(b) A 28-(b) A 48-(b) A 9-(b) A 29-(b) A 49-(b) A 10-(b) E 30-(b) A 50-(b) C 11-(b) A 31-(b) A 51-(b) C 12-(b) B 32-(b) A 52-(b) A 13-(b) A 33-(b) A 53-(b) A 14-(b) A 34-(b) A 54-(b) A 15-(b) A 35-(b) A 55-(b) A 16-(b) A 36-(b) A 56-(b) B 17-(b) B 37-(b) A 18-(b) A 38-(b) A 19-(b) A 39-(b) A 20-(b) A 40-(b) A
221102_G2572WO English Translation (Final) clean copy
[0455] (Test Example 3) TGF- activation inhibition test
In Test Example 3, the inhibition of TGF
activation by a compound was detected with the inhibition
of SMAD3 phosphorylation as an index. In Test Example 3,
the same reagents as in Test Example 1 were used unless
otherwise specified.
Human fibronectin was prepared at 14 pg/mL with PBS
and then dispensed at 50 pL/well to a 96-well plate. The
plate was left standing at room temperature for 1 hour or
longer, followed by the removal of the fibronectin
solution. The plate was washed with PBS and used as a
coated plate.
[0456] A test compound dissolved in DMSO was added to
RPMI medium 1640 (manufactured by Thermo Fisher
Scientific, Inc., 11875) containing 1% FBS and 1%
penicillin/streptomycin/amphotericin B to prepare a
medium containing the test compound having a
concentration of two times the final concentration. This
medium was added at 100 pL/well to the coated plate.
TFK-1 cells (Cell Resource Center for Biomedical
Research, Institute of Development, Aging and Cancer,
Tohoku University) cultured in RPMI medium 1640
containing 10% FBS and 1%
penicillin/streptomycin/amphotericin B were suspended in
RPMI medium 1640 containing 1% FBS and 1%
penicillin/streptomycin/amphotericin B to prepare a cell
suspension of 1 x 106 cells/mL. The prepared cell
221102_G2572WO English Translation (Final) clean copy
suspension was further added at 100 pL/well and cultured
for 2 days in a 5% C02 incubator (37°C).
[0457] Phosphorylated SMAD3 in the cells thus cultured
for 2 days was measured using Phospho-SMAD3 (Ser423/425)
cellular kit (manufactured by cisbio, 63ADK025), and a
DeltaF value was calculated according to the calculation
expression of the kit. Inhibition when 10 pM LY364947
(FUJIFILM Wako Pure Chemical Corp., 123-05981) was added
instead of the test compound was defined as 100%
inhibition, and inhibition when DMSO was added instead
was defined as 0% inhibition. The rate of inhibition of
phosphorylated SMAD3 by the test compound was calculated
according to the following expression.
Rate of inhibition of phosphorylated SMAD3 (%) = [1
- {(DeltaF value in the case of adding the test compound)
- (DeltaF value in the case of adding 10 pM LY364947)}
{(DeltaF value in the case of adding DMSO) - (DeltaF / value in the case of adding 10 pM LY364947)}] x 100
[0458] An IC 50 value was calculated from the rate of
inhibition of phosphorylated SMAD3 by the test compound.
The results about each test compound in this test are
shown in Table 20 below. A: IC50 < 10 nM, B: 10 nM < IC 50
< 50 nM, C: 50 nM < IC50 < 100 nM, or D: IC 50 > 100 nM.
221102_G2572WO English Translation (Final) clean copy
Table 20. Results of TGF-p activation inhibition test about compound of Example Example No. IC 50 Example No. IC 50 Example No. IC50 1-(b) A 21-(b) A 41-(b) A 2-(b) A 22-(b) B 42-(b) B 3-(b) A 23-(b) B 43-(b) A 4-(b) A 24-(b) A 44-(b) A -(b) B 25-(b) A 45-(b) B 6-(b) B 26-(b) A 46-(b) C 7-(b) A 27-(b) A 47-(b) B 8-(b) A 28-(b) B 48-(b) A 9-(b) C 29-(b) A 49-(b) A 10-(b) D 30-(b) A 50-(b) D 11-(b) A 31-(b) A 51-(b) C 12-(b) D 32-(b) A 52-(b) A 13-(b) B 33-(b) A 53-(b) A 14-(b) A 34-(b) A 54-(b) A 15-(b) A 35-(b) B 55-(b) A 16-(b) B 36-(b) B 56-(b) A 17-(b) A 37-(b) A 18-(b) B 38-(b) C 19-(b) A 39-(b) A 20-(b) A 40-(b) B
[0459] (Test Example 4) Active human hepatic stellate
cell dedifferentiation test
Human hepatic stellate cells (manufactured by
ScienCell Research Laboratories, Inc., 5300) were
suspended in DMEM containing 10% FBS and 1%
penicillin/streptomycin/amphotericin B to prepare a cell
suspension of 0.5 x 105 cells/mL, which was regarded as
cells at the start of differentiation. The cells at the
start of differentiation were dispensed at 1 mL/well to a
221102_G2572WO English Translation (Final) clean copy
12-well plate and cultured, for differentiation, for 3
days in a 5% C02 incubator (37°C).
The culture solution in each well was removed, and
DMEM containing 10% FBS and 1%
penicillin/streptomycin/amphotericin B supplemented with
a test compound dissolved in DMSO (final DMSO
concentration: 0.1%) was added at 1 mL/well. The
resultant was further cultured for 2 days in a 5% C02
incubator (37°C) . In Test Example 4, the same reagents
as in Test Example 1 were used unless otherwise
specified.
[0460] RNA in the cultured cells was extracted using
NucleoSpin(R) RNA (manufactured by Takara Bio Inc.,
U0955C), and cDNA was further synthesized from the
extracted RNA using Prime Script(TM) RT reagent Kit with
gDNA Eraser (manufactured by Takara Bio Inc., RR047A).
Real time PCR was performed using the synthesized cDNA,
various primers and TB Green(R) Premix Ex Taq(TM) II
(manufactured by Takara Bio Inc., RR820A) and using PCR
Thermal Cycler Dice Real Time (manufactured by Takara Bio
Inc., System Code TP910, Model Code TP900). The primers
used were actin alpha 2, smooth muscle (ACTA2) primer
(manufactured by Takara Bio Inc., HA136273) and
glyceraldehyde-3-phosphate dehydrogenase (GAPDH) primer
(manufactured by Takara Bio Inc., HA067812).
[0461] The amplification curve of PCR was analyzed by the
comparative Ct method. The expression level of ACTA2
221102_G2572WO English Translation (Final) clean copy
mRNA was corrected with the expression level of GAPDH
mRNA. An ACTA2 mRNA expression level when DMSO was added
instead of the test compound was defined as 100%, and an
ACTA2 mRNA expression level in the cells at the start of
differentiation was defined as 0%. The rate of decrease
in ACTA2 mRNA expression by the test compound was
calculated according to the following expression.
Rate of decrease in ACTA2 mRNA expression (%) = [1
{(ACTA2 mRNA expression level in the case of adding the
test compound) - (ACTA2 mRNA expression level in the
cells at the start of differentiation)} / {(ACTA2 mRNA
expression level in the case of adding DMSO) - (ACTA2
mRNA expression level in the cells at the start of
differentiation)}] x 100
[0462] An IC50 value was calculated from the rate of
decrease in ACTA2 mRNA expression by the test compound.
The results about each test compound in this test are
shown in Table 21 below. A: IC50 < 10 nM, B: 10 nM < IC50
< 50 nM, C: 50 nM < IC50 < 100 nM, or D: IC50 100 nM.
221102_G2572WO English Translation (Final) clean copy
Table 21. Results of active human hepatic stellate cell dedifferentiation test about compound of Example Example No. IC50 Example No. IC50 Example No. IC 50
1-(b) A 21-(b) B 41-(b) B 2-(b) B 42-(b) B 3-(b) B 43-(b) B 4-(b) B 24-(b) A -(b) A 25-(b) A 6-(b) B 26-(b) A 7-(b) A 8-(b) C 28-(b) B 48-(b) A 9-(b) B 49-(b) A 10-(b) D 30-(b) A 11-(b) A 31-(b) A 12-(b) B 32-(b) B 52-(b) B 13-(b) B 33-(b) B 53-(b) B 14-(b) A 34-(b) B 54-(b) B 15-(b) A 35-(b) A 55-(b) A 16-(b) A 36-(b) B 56-(b) A 17-(b) B 37-(b) B 18-(b) A 38-(b) A 19-(b) A 39-(b) A 20-(b) A 40-(b) B
[0463] (Test Example 5) Hepatic fibrosis suppression test
in hepatic fibrosis (non-alcoholic steatohepatitis: NASH)
model
Test Example 5 was carried out by partially
modifying the method of Matsumoto et al. (Int J Exp
Pathol., 94 (2), 93-103 (2013)).
C57BL/6J mice (male, Japan SLC, Inc.) were fed with
CDAHFD (choline-deficient amino acid-defined high fat
diet, supplemented with 0.1% methionine, 60 kcal% fat,
221102_G2572WO English Translation (Final) clean copy
manufactured by Research Diets Inc., A06071302) for 8
weeks to prepare hepatic fibrosis models. The start of
feeding with CDAHFD was defined as week 0. A test
compound or a vehicle was administered once a day during
a period from weeks 0 to 8. The group given the test
compound was used as a test compound administration
group, and the group given the vehicle was used as a
vehicle group.
Mice fed with CE-2 (CLEA Japan, Inc.) instead of
CDAHFD were used as a non-fibrotic group. Eight weeks
later, each mouse was euthanized by blood letting from
the abdomen under inhalation anesthesia with isoflurane,
and the liver was harvested.
Hydroxyproline (HYP) was quantified using a portion
of the liver, and a hepatic HYP level per unit weight was
calculated. The amount of increase in hepatic HYP level
per unit weight of the vehicle group based on the hepatic
HYP level of the non-fibrotic group was defined as 100%.
The rate of suppression of increase in HYP in the test
compound administration group was calculated.
As a result of testing compounds of Examples, the
compounds of Examples suppressed increase in HYP.
[0464] (Test Example 6) Transcellular transport test
using Caco2 cell
Caco2 cells (purchased from ATCC) were cultured in
DMEM medium (Thermo Fisher Scientific, Inc., 11965)
containing 10% FBS (Thermo Fisher Scientific, Inc.,
221102_G2572WO English Translation (Final) clean copy
10082) and 1% penicillin/streptomycin/amphotericin B
(Thermo Fisher Scientific, Inc., 15240). A transcellular
transport test was carried out in accordance with
MultiScreen Caco2 Drug Transport Assembly in a 96-well
system (Application Note AN1727EN, MultiScreen Caco-2
Assay System, Merck) by partially modifying it. The
Caco2 cells were inoculated at a cell density of 4 x 104
cells/well onto a micropore polycarbonate insert of a 96
well plate (Merck, Millcell-96 culture insert plate 0.4
pm pore size, well membrane area = 0.11 cm 2 , PSHT004S5)
and cultured for 20 to 22 days, and the resulting
monolayer was used in the transcellular transport test.
The medium of the inoculated cells was replaced with a
fresh one every 3 to 4 days.
[0465] Apical-to-basolateral (A-to-B) transport in the
Caco2 cell monolayer membrane was performed by using HBSS
(Thermo Fisher Scientific, Inc., 14025) medium (pH = 7.4)
containing 20 mM HEPES (Thermo Fisher Scientific, Inc.,
15630), and adding HEPES-containing HBSS medium
containing 10 pM compound (a 10 mM solution of the
compound in DMSO was added at a 1000-fold dilution; final
DMSO concentration: 0.1%) at 75 pL/well to the apical
side and HEPES-containing HBSS medium supplemented with
no compound at 250 pL/well to the basolateral side,
followed by culture at 370C for 90 minutes. After
culture, a 100 pL aliquot was collected from the
basolateral side, and the concentration of a permeated
221102_G2572WO English Translation (Final) clean copy
compound was measured by LC-MS/MS under measurement
conditions mentioned later. Papp (apparent permeability;
cm/sec) was calculated from the determined concentration.
Papp was determined according to the expression given
below by calculating the slope (dA/dt; nmol/sec) of a
straight line drawn between two points 0 minutes and zero
amount of the compound in a graph plotted with an assay
time (90 min) on the abscissa against the amount of the
compound in the solution on the permeated side on the
ordinate. As for an assay concentration, the
concentration in the compound in the HEPES-containing
HBSS medium containing the added compound was measured by
UV-HPLC under measurement conditions mentioned later.
Papp (cm/sec) = CO x S x (dA / dt)
CO: assay concentration, S: plate membrane area =
0.11 cm 2
[0466] <Measurement conditions of LC-MS/MS analysis>
LC: LC20 or LC30 HPLC system manufactured by Shimadzu
Corp.
Column: Phenomenex Kinetex C18 (50 mm x 2.1 mm, 2.6
Pm)
Column temperature: 40 0 C
Flow rate: 0.3 mL/min
Mobile phase A: 0.1% aqueous formic acid solution
Mobile phase B: 0.1% formic acid, 50%
acetonitrile/methanol mixed solution
221102_G2572WO English Translation (Final) clean copy
Gradient: 0-2 min; A/B = 90/10 to 10/90, 2-3 min;
A/B = 10/90, 3-3.01 min; A/B = 10/90 to 90/10
MS: Q-Trap3200 manufactured by AB Sciex Pte. Ltd.
Ionization: ESI
Mode: Positive
[0467] <Measurement conditions of UV-HPLC analysis>
LC: LC20 or LC30 HPLC system manufactured by Shimadzu
Corp.
Column: Phenomenex Kinetex C18 (100 mm x 2.1 mm, 2.6
pm)
Column temperature: 40 0 C
Flow rate: 0.25 mL/min
Mobile phase A: 0.1% aqueous formic acid solution
Mobile phase B: 0.1% formic acid, 50%
acetonitrile/methanol mixed solution
Gradient: 0-3 min; A/B = 90/10, 3-11 min; A/B =
/10 to 5/95, 11-15 min; A/B = 5/95, 15-15.1 min; A/B =
/95 to 90/10
UV detector: photodiode array detector
[0468] The results about each test compound in this test
are shown in Table 22 below. A: Papp > 1 x 10-6 (cm/sec),
B: 1 x 10-6 (cm/sec) > Papp > 5 x 10-7 (cm/sec), C: 5 x 10
? (cm/sec) > Papp > 1 x 10-7 (cm/sec), or D: Papp < 1 x
-7 (cm/sec)
221102_G2572WO English Translation (Final) clean copy
Table 22. Results of transcellular transport test using Caco2 cell about compound of Example Example No. Papp Example No. Papp Example No. Papp 1-(b) A 21-(b) A 41-(b) A 2-(b) B 22-(b) A 42-(b) A 3-(b) A 23-(b) A 43-(b) A 4-(b) A 24-(b) B 44-(b) B -(b) B 25-(b) A 45-(b) B 6-(b) A 26-(b) A 46-(b) A 7-(b) B 27-(b) A 47-(b) A 8-(b) B 28-(b) A 48-(b) A 9-(b) A 29-(b) C 49-(b) B 10-(b) A 30-(b) B 50-(b) A 11-(b) A 31-(b) A 51-(b) A 12-(b) A 32-(b) A 52-(b) C 13-(b) A 33-(b) A 53-(b) C 14-(b) A 34-(b) A 54-(b) C 15-(b) A 35-(b) A 55-(b) C 16-(b) A 36-(b) A 56-(b) C 17-(b) A 37-(b) A 18-(b) A 38-(b) A 19-(b) A 39-(b) A 20-(b) A 40-(b) A
[0469] (Test Example 7) Dansyl glutathione (dGSH)
trapping assay
Among metabolites generated from a test compound
through metabolism in liver microsomes, a metabolite that
reacted with dansyl glutathione (dGSH) was detected and
quantified as a method for detecting a reactive
metabolite. The concentration of the metabolite-dGSH
conjugate was measured using fluorescence detection UPLC
system (manufactured by Shimadzu Corp.).
221102_G2572WO English Translation (Final) clean copy
As a result of testing compounds of Examples, the
compounds of Examples generated no or few dGSH adducts.
This demonstrated the compound represented by the general
formula (I) or (II) of the present invention or a
pharmaceutically acceptable salt thereof is a compound
excellent in safety that generates no or few reactive
metabolites.
[0470] (Test Example 8) Hepatic fibrosis suppression test
in carbon tetrachloride-induced hepatic fibrosis model
Carbon tetrachloride diluted 2-fold with olive oil
was subcutaneously administered at 2 mL/kg twice a week
up to the completion of a test to the back of C57BL/6J
mice (female, Charles River Laboratories Japan, Inc.)
under inhalation anesthesia with isoflurane to prepare
carbon tetrachloride-induced hepatic fibrosis models. A
test compound was orally administered once a day for 56
days from the start of model preparation. Then, the
liver was collected, and HYP in the left lateral hepatic
lobe was quantified.
As a result of testing compounds of Examples, the
compounds of Examples suppressed increase in HYP.
[0471] (Test Example 9) Pulmonary fibrosis suppression
test in bleomycin-induced idiopathic pulmonary fibrosis
model
Bleomycin (manufactured by Nippon Kayaku Co., Ltd.)
diluted with physiological saline was transtracheally
administered once at 3.0 mg/kg (50 pL/animal) to C57BL/6J
221102_G2572WO English Translation (Final) clean copy
mice (female, Japan SLC, Inc.) under triple anesthesia
(medetomidine hydrochloride/midazolam/butorphanol
tartrate) using Microsprayer (manufactured by Penn
Century, Inc.) to prepare bleomycin-induced idiopathic
pulmonary fibrosis models. A test compound was
administered thereto for 14 days from 7 days after
bleomycin administration. Then, the lung was collected,
and HYP was quantified using the pulmonary left lobe.
As a result of testing compounds of Examples, the
compounds of Examples suppressed increase in HYP.
[0472] (Test Example 10) Renal fibrosis suppression test
in unilateral ureteral obstruction model with renal
fibrosis
Test Example 10 was performed with reference to the
method of Swaney et al. (The Journal of Pharmacology and
Experimental Therapeutics 336 (3): 693-700 (2011)). The
left ureter of C57BL/6J mice (male, Japan SLC, Inc.) was
ligated with a suture thread under inhalation anesthesia
with isoflurane to prepare unilateral ureteral
obstruction models with renal fibrosis. A test compound
was administered thereto for 10 days from model
preparation. Then, the left kidney was collected, and
HYP was quantified.
As a result of testing compounds of Examples, the
compounds of Examples suppressed increase in HYP.

Claims (16)

  1. Claims
    [Claim 1]
    A compound represented by the following general
    formula (I) or (II) or a pharmaceutically acceptable salt
    thereof:
    Y Y
    HN N HN N O H Y H HN N CO2R HN N CO2R I / 0 A O 0 A R 1/: R1 N-NH HN-N
    (I) (II) wherein
    A is a C 6 -C1 0 aryl group or a heteroaryl group,
    wherein at least one hydrogen atom of the aryl group or
    the heteroaryl group is optionally replaced with a
    substituent selected from the group consisting of a
    halogen atom, a hydroxy group, a C 1 -C6 alkyl group, a C1
    C 6 haloalkyl group, a C 2 -C 6 alkenyl group, a C 2 -C 6 alkynyl
    group, a C 1 -C 6 alkoxy group, a C 1 -C 6 haloalkoxy group, a
    C 3 -C 6 cycloalkyl group, a C 3 -C 6 cycloalkenyl group, a C 3 -C 6
    cycloalkoxy group, a heterocyclyl group, a heteroaryl
    group optionally substituted by a C 1 -C 6 alkyl group, a
    cyano group, a carboxyl group, a carbamoyl group, a C 1 -C6
    alkoxycarbonyl group, a C1 -C 6 alkylsulfanyl group, and a
    C1 -C 6 alkylsulfonyl group,
    R is a hydrogen atom or a C 1 -C6 alkyl group,
    R' is a hydrogen atom or a halogen atom, and
    Y is a hydrogen atom, a fluorine atom or a hydroxy
    group.
  2. [Claim 2]
    A compound represented by the following general
    formula (I) or a pharmaceutically acceptable salt
    thereof:
    Y
    HN N HN O N N CO 2 R
    R 1 1 H0 A N-NH
    (I) wherein
    A is a C6-C10 aryl group or a heteroaryl group,
    wherein at least one hydrogen atom of the aryl group or
    the heteroaryl group is optionally replaced with a
    substituent selected from the group consisting of a
    halogen atom, a hydroxy group, a C1-C6 alkyl group, a C1
    C6 haloalkyl group, a C2-C6 alkenyl group, a C2-C6 alkynyl
    group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a
    C3-C6 cycloalkyl group, a C3-C6 cycloalkenyl group, a C3-C6
    cycloalkoxy group, a heterocyclyl group, a heteroaryl
    group optionally substituted by a C1-C6 alkyl group, a
    cyano group, a carboxyl group, a carbamoyl group, a C1-C6
    alkoxycarbonyl group, a C1-C6 alkylsulfanyl group, and a
    C1-C6 alkylsulfonyl group,
    R is a hydrogen atom or a C1-C6 alkyl group,
    R' is a hydrogen atom or a halogen atom, and
    Y is a hydrogen atom, a fluorine atom or a hydroxy
    group.
  3. [Claim 3]
    A compound represented by the following general
    formula (II) or a pharmaceutically acceptable salt
    thereof:
    Y
    HN N HN N CO2R 0 O A R1 HN-N
    (10)
    wherein
    A is a C6-C1O aryl group or a heteroaryl group,
    wherein at least one hydrogen atom of the aryl group or
    the heteroaryl group is optionally replaced with a
    substituent selected from the group consisting of a
    halogen atom, a hydroxy group, a C1-C6 alkyl group, a C1
    C6 haloalkyl group, a C2-C6 alkenyl group, a C2-C6 alkynyl
    group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a
    C3-C6 cycloalkyl group, a C3-C6 cycloalkenyl group, a C3-C6
    cycloalkoxy group, a heterocyclyl group, a heteroaryl
    group optionally substituted by a C1-C6 alkyl group, a
    cyano group, a carboxyl group, a carbamoyl group, a C1-C6 alkoxycarbonyl group, a C1-C6 alkylsulfanyl group, and a
    C1-C6 alkylsulfonyl group,
    R is a hydrogen atom or a C1-C alkyl group,
    R' is a hydrogen atom or a halogen atom, and
    Y is a hydrogen atom, a fluorine atom or a hydroxy
    group.
  4. [Claim 4]
    The compound according to any one of claims 1 to 3
    or a pharmaceutically acceptable salt thereof, wherein A
    is a phenyl group, wherein
    at least one hydrogen atom of the phenyl group is
    optionally replaced with a substituent selected from the
    group consisting of a halogen atom, a hydroxy group, a
    C1-C6 alkyl group, a C1-C6 haloalkyl group, a C2-C6 alkenyl
    group, a C2-C6 alkynyl group, a C1-C6 alkoxy group, a C1-C6
    haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
    cycloalkenyl group, a C3-C6 cycloalkoxy group, a
    heterocyclyl group, a heteroaryl group optionally
    substituted by a C1-C6 alkyl group, a cyano group, a
    carboxyl group, a carbamoyl group, a C1-C6 alkoxycarbonyl
    group, a C1-C6 alkylsulfanyl group, and a C1-C6
    alkylsulfonyl group.
  5. [Claim 5]
    The compound according to any one of claims 1 to 4
    or a pharmaceutically acceptable salt thereof, wherein A
    is a phenyl group, wherein at least one hydrogen atom of the phenyl group is optionally replaced with a substituent selected from the group consisting of a halogen atom, a C1-C6 alkyl group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a C1-C6 haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6 cycloalkoxy group, a heterocyclyl group, and a heteroaryl group optionally substituted by a C1-C6 alkyl group.
  6. [Claim 6]
    The compound according to any one of claims 1 to 5
    or a pharmaceutically acceptable salt thereof, wherein Y
    is a fluorine atom.
  7. [Claim 7]
    The compound according to any one of claims 1 to 6
    or a pharmaceutically acceptable salt thereof, wherein A
    is a group represented by the following formula (i):
    RS6 R2
    R5 / R3 R4
    0) wherein
    R 2 is a hydrogen atom or a halogen atom,
    R 3 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
    group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
    C1-C6 haloalkoxy group, a C3-C6 cycloalkyl group, a C3-C6
    cycloalkoxy group, a heterocyclyl group, or a heteroaryl
    group optionally substituted by a C1-C6 alkyl group,
    R4 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
    group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, or a
    C1-C6 haloalkoxy group,
    R5 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
    group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
    C1-C6 haloalkoxy group, or a heteroaryl group optionally
    substituted by a C1-C6 alkyl group, and
    R 6 is a hydrogen atom or a halogen atom.
  8. [Claim 8]
    The compound according to claim 7 or a
    pharmaceutically acceptable salt thereof, wherein
    R 2 is a hydrogen atom,
    R 3 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
    group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, a
    C1-C6 haloalkoxy group, or a heteroaryl group optionally
    substituted by a C1-C6 alkyl group,
    R 4 is a hydrogen atom or a halogen atom,
    R5 is a hydrogen atom, a halogen atom, a C1-C6 alkyl
    group, a C1-C6 haloalkyl group, a C1-C6 alkoxy group, or a
    C1-C6 haloalkoxy group, and
    R 6 is a hydrogen atom or a halogen atom.
  9. [Claim 9]
    The compound according to claim 1 or a
    pharmaceutically acceptable salt thereof, wherein the
    compound is selected from the group consisting of
    (3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-bromo-5-(trifluoromethyl)phenyl)-3-(2-(4
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
    iodo-5-(trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
    methyl-5-(trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
    methoxy-5-(trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
    fluoro-5-(trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(3,5-bis(trifluoromethyl)phenyl)-3-(2-(4-((5
    fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-(1H-pyrazol-1-yl)-5
    (trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
    tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(2
    fluoro-3-(trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(2-chloro-3-(trifluoromethyl)phenyl)-3-(2-(4
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(2
    fluoro-5-(trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(2-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
    fluoro-3-(trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
    methyl-3-(trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(3-chloro-4-fluoro-5
    (trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
    tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-bromo-4-methoxyphenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3,5-dichlorophenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-bromo-5-chlorophenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3,5-dibromophenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-bromo-5-iodophenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-chloro-5-iodophenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(5-chloro-2-fluorophenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(5-bromo-2-fluorophenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-(difluoromethoxy)-5
    (trifluoromethyl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
    tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-chloro-5-(difluoromethoxy)phenyl)-3-(2-(4
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-bromo-5-(difluoromethoxy)phenyl)-3-(2-(4
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-(difluoromethoxy)-4-fluorophenyl)-3-(2-(4
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(5-(difluoromethoxy)-2-fluorophenyl)-3-(2-(4
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-(difluoromethoxy)-5-(1H-pyrazol-1
    yl)phenyl)-3-(2-(4-((5-fluoro-1,4,5,6
    tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3,5-bis(difluoromethoxy)phenyl)-3-(2-(4-((5
    fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-chloro-5-(trifluoromethoxy)phenyl)-3-(2
    (4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-bromo-5-(trifluoromethoxy)phenyl)-3-(2-(4
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(2
    fluoro-5-(trifluoromethoxy)phenyl)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
    fluoro-3-(trifluoromethoxy)phenyl)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
    (trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
    (trifluoromethoxy)phenyl)propanoic acid,
    (3S)-3-(3-(difluoromethoxy)phenyl)-3-(2-(4-((5
    fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-(difluoromethyl)phenyl)-3-(2-(4-((5
    fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-cyclopropylphenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-cyclopropoxyphenyl)-3-(2-(4-((5-fluoro
    1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-chlorophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
    tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-bromophenyl)-3-(2-(4-((5-fluoro-1,4,5,6
    tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-(1H-pyrazol-1-yl)phenyl)-3-(2-(4-((5
    fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-(3,5-dimethyl-1H-pyrazol-1-yl)phenyl)-3
    (2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)
    1H-indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(3
    morpholinophenyl)propanoic acid,
    (3S)-3-(4-(difluoromethoxy)phenyl)-3-(2-(4-((5
    fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (3S)-3-(2-(4-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-6-carboxamido)acetamido)-3-(4
    (trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(3
    fluoro-4-((5-fluoro-1,4,5,6-tetrahydropyrimidin-2
    yl)amino)-1H-indazole-6-carboxamido)acetamido)propanoic
    acid,
    (3S)-3-(2-(3-chloro-4-((5-fluoro-1,4,5,6
    tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)-3-(3-chloro-5
    (trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
    ((5-hydroxy-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-6-carboxamido)acetamido)propanoic acid,
    (S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(4
    ((1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H-indazole-6
    carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-chloro-5-(trifluoromethyl)phenyl)-3-(2-(6
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-4-carboxamido)acetamido)propanoic acid,
    (3S)-3-(3-bromo-5-(trifluoromethyl)phenyl)-3-(2-(6
    ((5-fluoro-1,4,5,6-tetrahydropyrimidin-2-yl)amino)-1H
    indazole-4-carboxamido)acetamido)propanoic acid,
    (3S)-3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-4-carboxamido)acetamido)-3-(3
    iodo-5-(trifluoromethyl)phenyl)propanoic acid,
    (3S)-3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-4-carboxamido)acetamido)-3-(4
    fluoro-3-(trifluoromethyl)phenyl)propanoic acid, and
    (3S)-3-(2-(6-((5-fluoro-1,4,5,6-tetrahydropyrimidin
    2-yl)amino)-1H-indazole-4-carboxamido)acetamido)-3-(3
    (trifluoromethyl)phenyl)propanoic acid.
  10. [Claim 10]
    A pharmaceutical composition comprising a compound
    according to any one of claims 1 to 9 or a
    pharmaceutically acceptable salt thereof.
  11. [Claim 11]
    The pharmaceutical composition according to claim 10
    for preventing, alleviating and/or treating a disease
    related to integrin av or a disease that is improved by
    inhibiting integrin av.
  12. [Claim 12]
    The pharmaceutical composition according to claim 9
    or 10 for the remedy of fibrosis.
  13. [Claim 13]
    A method for preventing, alleviating and/or treating
    a disease related to integrin av or a disease that is
    improved by inhibiting integrin av, comprising
    administering a compound according to any one of claims 1
    to 9 or a pharmaceutically acceptable salt thereof to a
    subject.
  14. [Claim 14]
    A method for treating fibrosis, comprising
    administering a compound according to any one of claims 1
    to 9 or a pharmaceutically acceptable salt thereof to a
    subject.
  15. [Claim 15]
    The compound according to any one of claims 1 to 9
    or a pharmaceutically acceptable salt thereof for use in
    the prevention, alleviation and/or remedy of a disease
    related to integrin av or a disease that is improved by
    inhibiting integrin av.
  16. [Claim 16]
    The compound according to any one of claims 1 to 9
    or a pharmaceutically acceptable salt thereof for use in
    the remedy of fibrosis.
AU2021270095A 2020-05-14 2021-05-13 1, 4, 5, 6-tetrahydropyrimidine-2-amine derivative Pending AU2021270095A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
JP2020085420 2020-05-14
JP2020-085420 2020-05-14
PCT/JP2021/018252 WO2021230325A1 (en) 2020-05-14 2021-05-13 1, 4, 5, 6-tetrahydropyrimidine-2-amine derivative

Publications (1)

Publication Number Publication Date
AU2021270095A1 true AU2021270095A1 (en) 2022-12-22

Family

ID=78524455

Family Applications (1)

Application Number Title Priority Date Filing Date
AU2021270095A Pending AU2021270095A1 (en) 2020-05-14 2021-05-13 1, 4, 5, 6-tetrahydropyrimidine-2-amine derivative

Country Status (10)

Country Link
US (1) US20230192661A1 (en)
EP (1) EP4151632A4 (en)
JP (1) JPWO2021230325A1 (en)
KR (1) KR20230011967A (en)
CN (1) CN115551846B (en)
AU (1) AU2021270095A1 (en)
BR (1) BR112022023058A2 (en)
CA (1) CA3182964A1 (en)
MX (1) MX2022014200A (en)
WO (1) WO2021230325A1 (en)

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2023085396A1 (en) * 2021-11-12 2023-05-19 Ube株式会社 Pharmaceutical composition for providing treatment for or preventing alport syndrome

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
ES2161373T3 (en) 1995-08-30 2001-12-01 Searle & Co DERIVATIVES OF META-GUANIDINA, UREA, THIOUREA OR AZACICLICO-AMINOBENZOIC ACID AS INTEGRINE ANTAGONISTS.
ZA994406B (en) 1998-03-04 2000-02-11 Searle & Co Meta-azacyclic amino benzoic acid and derivatives thereof.
US6372719B1 (en) 1998-03-04 2002-04-16 Jay Cunningham ανβ3 integrin antagonists in combination with chemotherapeutic agents
JP2002511462A (en) * 1998-04-10 2002-04-16 ジー・ディー・サール・アンド・カンパニー Heterocyclic glycyl β-alanine derivatives as vitronectin antagonists
MXPA05006732A (en) * 2002-12-20 2005-09-08 Pharmacia Corp The r-isomer of beta amino acid compounds as integrin receptor antagonists derivatives.
US8716226B2 (en) 2012-07-18 2014-05-06 Saint Louis University 3,5 phenyl-substituted beta amino acid derivatives as integrin antagonists
AU2013292580B2 (en) * 2012-07-18 2017-07-20 Saint Louis University Beta amino acid derivatives as integrin antagonists
RU2729518C2 (en) 2015-12-30 2020-08-07 Сент-Луис Юниверсити Meta-azacyclic aminobenzoic acid derivatives as pan-integrin antagonists

Also Published As

Publication number Publication date
WO2021230325A1 (en) 2021-11-18
CA3182964A1 (en) 2021-11-18
JPWO2021230325A1 (en) 2021-11-18
CN115551846A (en) 2022-12-30
MX2022014200A (en) 2022-12-07
BR112022023058A2 (en) 2022-12-20
EP4151632A4 (en) 2024-04-24
KR20230011967A (en) 2023-01-25
EP4151632A1 (en) 2023-03-22
US20230192661A1 (en) 2023-06-22
CN115551846B (en) 2024-03-08

Similar Documents

Publication Publication Date Title
US20230271960A1 (en) Amino acid compounds and methods of use
CA3038479C (en) Pyrazolopyridine derivative having glp-1 receptor agonist effect
US9278968B2 (en) Imidazopyridine compounds
AU2010222589B2 (en) Benzofuranyl derivatives used as glucokinase inhibitors
JP6242390B2 (en) 2-Aminopyrazine derivatives as CSF-1R kinase inhibitors
CA3136745A1 (en) Dosage forms and regimens for amino acid compounds
JP2013545740A (en) Substituted heteroaromatic carboxamide derivatives and urea derivatives as vanilloid receptor ligands
JP7461104B2 (en) Pharmaceutical composition containing pyrazolopyridine derivative having GLP-1 receptor agonist activity
WO2007055418A1 (en) Aza-substituted spiro derivative
PT1024138E (en) Pyrazole derivatives
BRPI1011043B1 (en) phenylureas and substituted phenylamides as vanyloid receptor ligands, their uses and their preparation process, and pharmaceutical composition
PL188077B1 (en) Substituted 1-phenypyrazole-3-carboxyamides as active substances in respect to neurotensin rceptors, their production and pharmaceutic compositions containing them
CA2681861A1 (en) Ornithine derivative
JP2008545756A (en) 1-methyl-1H-pyrazole-4-carboxamides useful as cancer chemotherapeutic agents
EP4058458A2 (en) Novel functionalized lactones as modulators of the 5-hydroxytryptamine receptor 7 and their method of use
AU2021270095A1 (en) 1, 4, 5, 6-tetrahydropyrimidine-2-amine derivative
WO2010023512A1 (en) Novel vanilloid receptor modulators, process for their preparation and pharmaceutical compositions containing them
US20100184777A1 (en) Substituted Pyrazinone Amides
JP5647379B2 (en) Quinolinyl glucagon receptor modulator
CA2700936A1 (en) Production method for pyrazol-3-yl-benzamide derivative
WO2023085396A1 (en) Pharmaceutical composition for providing treatment for or preventing alport syndrome
US20230286948A1 (en) Haloalkylpyridyl triazole mll1-wdr5 protein-protein interaction inhibitor
NZ284731A (en) Cyclobut-3-ene-1,2-dione derivatives as smooth muscle relaxants
JP2013542231A (en) Substituted bicyclic aromatic carboxamide derivatives and urea derivatives as vanilloid receptor ligands
NZ751725B2 (en) Pyrazolopyridine derivative having glp-1 receptor agonist effect